middle east respiratory syndrome and severe acute ... · the outbreaks of middle east respiratory...

32
REVIEW ARTICLE Middle East Respiratory Syndrome and Severe Acute Respiratory Syndrome: Current Therapeutic Options and Potential Targets for Novel Therapies Julie Dyall 1 Robin Gross 1 Jason Kindrachuk 2 Reed F. Johnson 3 Gene G. Olinger Jr. 4 Lisa E. Hensley 1 Matthew B. Frieman 5 Peter B. Jahrling 1,3 Published online: 15 November 2017 Ó Springer International Publishing AG 2017 Abstract No specific antivirals are currently available for two emerging infectious diseases, Middle East respiratory syndrome (MERS) and severe acute respiratory syndrome (SARS). A literature search was performed covering pathogenesis, clinical features and therapeutics, clinically developed drugs for repurposing and novel drug targets. This review presents current knowledge on the epidemi- ology, pathogenesis and clinical features of the SARS and MERS coronaviruses. The rationale for and outcomes with treatments used for SARS and MERS is discussed. The main focus of the review is on drug development and the potential that drugs approved for other indications provide for repurposing. The drugs we discuss belong to a wide range of different drug classes, such as cancer therapeutics, antipsychotics, and antimalarials. In addition to their activity against MERS and SARS coronaviruses, many of these approved drugs have broad-spectrum potential and have already been in clinical use for treating other viral infections. A wealth of knowledge is available for these drugs. However, the information in this review is not meant to guide clinical decisions, and any therapeutic described here should only be used in context of a clinical trial. Potential targets for novel antivirals and antibodies are discussed as well as lessons learned from treatment development for other RNA viruses. The article concludes with a discussion of the gaps in our knowledge and areas for future research on emerging coronaviruses. Key Points The outbreaks of Middle East respiratory syndrome (MERS) and severe acute respiratory syndrome (SARS) were caused by emerging coronaviruses. A variety of approaches for developing therapeutics are discussed with emphasis on drugs that have been approved for other indications and could be repurposed for treating emerging coronaviral infections. The recent MERS and SARS outbreaks highlight the importance of a panel of well-characterized broad- spectrum antivirals for treating emerging viral infections 1 Introduction An electronic literature search for countermeasures against Middle East respiratory syndrome coronavirus (MERS- CoV) and severe acute respiratory syndrome coronavirus (SARS-CoV) was performed using PubMed and Google & Julie Dyall [email protected] 1 Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, USA 2 Department of Medical Microbiology, University of Manitoba, Winnipeg, MN, Canada 3 Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, USA 4 University of Boston, Boston, MA, USA 5 Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, MD, USA Drugs (2017) 77:1935–1966 https://doi.org/10.1007/s40265-017-0830-1

Upload: others

Post on 20-Aug-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Middle East Respiratory Syndrome and Severe Acute ... · The outbreaks of Middle East respiratory syndrome (MERS) and severe acute respiratory syndrome (SARS) were caused by emerging

REVIEW ARTICLE

Middle East Respiratory Syndrome and Severe Acute RespiratorySyndrome: Current Therapeutic Options and Potential Targetsfor Novel Therapies

Julie Dyall1 • Robin Gross1 • Jason Kindrachuk2 • Reed F. Johnson3 •

Gene G. Olinger Jr.4 • Lisa E. Hensley1 • Matthew B. Frieman5 • Peter B. Jahrling1,3

Published online: 15 November 2017

� Springer International Publishing AG 2017

Abstract No specific antivirals are currently available for

two emerging infectious diseases, Middle East respiratory

syndrome (MERS) and severe acute respiratory syndrome

(SARS). A literature search was performed covering

pathogenesis, clinical features and therapeutics, clinically

developed drugs for repurposing and novel drug targets.

This review presents current knowledge on the epidemi-

ology, pathogenesis and clinical features of the SARS and

MERS coronaviruses. The rationale for and outcomes with

treatments used for SARS and MERS is discussed. The

main focus of the review is on drug development and the

potential that drugs approved for other indications provide

for repurposing. The drugs we discuss belong to a wide

range of different drug classes, such as cancer therapeutics,

antipsychotics, and antimalarials. In addition to their

activity against MERS and SARS coronaviruses, many of

these approved drugs have broad-spectrum potential and

have already been in clinical use for treating other viral

infections. A wealth of knowledge is available for these

drugs. However, the information in this review is not meant

to guide clinical decisions, and any therapeutic described

here should only be used in context of a clinical trial.

Potential targets for novel antivirals and antibodies are

discussed as well as lessons learned from treatment

development for other RNA viruses. The article concludes

with a discussion of the gaps in our knowledge and areas

for future research on emerging coronaviruses.

Key Points

The outbreaks of Middle East respiratory syndrome

(MERS) and severe acute respiratory syndrome

(SARS) were caused by emerging coronaviruses.

A variety of approaches for developing therapeutics

are discussed with emphasis on drugs that have been

approved for other indications and could be

repurposed for treating emerging coronaviral

infections.

The recent MERS and SARS outbreaks highlight the

importance of a panel of well-characterized broad-

spectrum antivirals for treating emerging viral

infections

1 Introduction

An electronic literature search for countermeasures against

Middle East respiratory syndrome coronavirus (MERS-

CoV) and severe acute respiratory syndrome coronavirus

(SARS-CoV) was performed using PubMed and Google

& Julie Dyall

[email protected]

1 Integrated Research Facility, Division of Clinical Research,

National Institute of Allergy and Infectious Diseases,

National Institutes of Health, Frederick, MD, USA

2 Department of Medical Microbiology, University of

Manitoba, Winnipeg, MN, Canada

3 Emerging Viral Pathogens Section, National Institute of

Allergy and Infectious Diseases, National Institutes of

Health, Frederick, MD, USA

4 University of Boston, Boston, MA, USA

5 Department of Microbiology and Immunology, University of

Maryland, School of Medicine, Baltimore, MD, USA

Drugs (2017) 77:1935–1966

https://doi.org/10.1007/s40265-017-0830-1

Page 2: Middle East Respiratory Syndrome and Severe Acute ... · The outbreaks of Middle East respiratory syndrome (MERS) and severe acute respiratory syndrome (SARS) were caused by emerging

Scholar from 2000 through April 17, 2017. The search (key

words: Middle East Respiratory Syndrome, Severe Acute

Respiratory Syndrome, inhibitors, antivirals, therapeutics,

FDA-approved) produced 1677 citations. References

selected discussed (1) pathogenesis and history of disease,

(2) clinical countermeasures used during the 2003 SARS

and 2012 MERS outbreaks and outcomes, and (3) the

efficacy of countermeasures targeting viral components

and cellular targets of MERS-CoV and SARS-CoV. The

main emphasis was on references for drug repurposing as

an alternative to the costly development of novel drugs for

emerging coronaviral infections.

1.1 Epidemiology of MERS and SARS

Since 2003, two human coronaviruses, SARS-CoV and

MERS-CoV, emerged as global public health threats.

SARS-CoV was first identified in February 2003 in

Guangdong Province, Peoples Republic of China and was

transmitted to humans from infected civets, likely infected

from bats [1, 2]. SARS-CoV spread to 29 additional

countries and was associated with high morbidity in

humans (e.g. atypical pneumonia). Ultimately, SARS was

contained in 2004 following a highly effective public

health response but resulted in 8098 confirmed cases and

774 deaths (Fig. 1a) [3]. In 2012, MERS emerged in The

Kingdom of Saudi Arabia and presented as a severe res-

piratory disease, with frequent gastrointestinal and renal

complications. MERS-CoV, the causative agent of MERS,

was later identified as a coronavirus. MERS-CoV has

subsequently spread to 27 additional countries (Fig. 1B)

[4]. As of September 12, 2017, 2080 confirmed cases of

MERS and 722 deaths were reported [5].

Coronaviruses are enveloped, single-stranded, positive-

sense RNA viruses (Fig. 2). They are members of the

Coronavirinae subfamily of viruses and together with the

Torovirinae subfamily comprise the Coronaviridae virus

family (order Nidovirales). Coronavirinae is divided into

four genera: alpha coronavirus, beta coronavirus, gamma

coronavirus, and delta coronavirus. The coronaviruses

share a similar genome organization. The open reading

frame 1a and 1b comprise nearly 2/3 of the genome and

encode the nonstructural proteins. The multiple structural

proteins, including spike, nucleocapsid, envelope, and

membrane proteins are encoded by downstream open

reading frames (Fig. 2) [6–8]. SARS-CoV and MERS-CoV

belong to the beta coronavirus genus. However, SARS-

CoV belongs to lineage B, and MERS-CoV belongs to

lineage C along with bat coronaviruses HKU4 and HKU5.

As MERS-CoV and bat coronaviruses are part of lineage C

and MERS-CoV RNA was found in a bat sample in The

Kingdom of Saudi Arabia, researchers hypothesize that

bats may be a natural reservoir for MERS-CoV [9, 10].

Results from a recent study support that bats may be a

reservoir for MERS-CoV; however, camels and goats are

thought to be intermediate hosts [11]. In this study, MERS-

CoV was isolated from nasal secretions of MERS-CoV-

infected dromedary camels that had a short, mild disease

progression.

The suspected reservoir for SARS-CoV is the Chinese

horseshoe bat [2]. However, the mechanism of emergence

and adaptation to make the virus zoonotic is still not def-

initely understood [2]. SARS-CoV-like isolates from these

bats have up to 95% sequence similarity to human and

civet SARS-CoV. During the initial outbreak, SARS-CoV

was originally isolated from palm civets found in a Chinese

market; but, SARS-CoV was not found in the wild palm

civet population [12]. Bats harbor many coronaviruses and

are considered the main reservoir for later infections in an

intermediate host, such as civets or camels, which spread

the disease to humans [2]. Human-to-human transmission

has been most commonly associated with health-care

workers and those with close, unprotected contact with

infected patients [13, 14].

1.2 Clinical Features

The clinical features of MERS and SARS are similar and

can range from asymptomatic or mild disease to severe

pneumonia with acute respiratory distress syndrome

(ARDS) and multi-organ failure [15]. Although MERS and

SARS are clinically similar, the MERS mortality rate is

40% and SARS’s mortality rate is 10% [16]. Approxi-

mately 75% of MERS cases were associated with under-

lying comorbidities with a 60% mortality rate in this

subgroup (including cardiopulmonary abnormalities, obe-

sity, and diabetes). In contrast, 10–30% of patients with

SARS have comorbidities with a mortality rate of 46%

within this subgroup [15, 16].

The development of symptomatic MERS and SARS

mostly occurs in adults (median age of 50 years; 40 years

for SARS). MERS and SARS symptoms typically follow a

mean incubation time of * 5 days (range 2–13 and

2–14 days, respectively) and include fever, chills, cough

(some associated with blood), shortness of breath, myalgia,

headache, nausea, vomiting, diarrhea, sore throat, and

malaise [15–17]. Progression from mild to severe disease is

more rapid with MERS as compared to SARS with means

of 7 and 11 days, respectively [15]. Secondary bacterial

infections have occurred in patients with severe MERS;

however, the role of these coinfections in MERS patho-

genesis has yet to be determined [18–20]. Laboratory

abnormalities associated with MERS and SARS patients

include elevated lactate dehydrogenase, elevated liver

enzymes; thrombocytopenia; lymphopenia and leukopenia

[21–23].

1936 J. Dyall et al.

Page 3: Middle East Respiratory Syndrome and Severe Acute ... · The outbreaks of Middle East respiratory syndrome (MERS) and severe acute respiratory syndrome (SARS) were caused by emerging

Radiographic abnormalities consistent with viral pneu-

monitis and ARDS are common in MERS and SARS.

Radiographic progression in the lower lobes has been

reported to be more rapid for MERS than SARS [21–23].

For SARS, disease in the lower lobes mimics pneumonia,

radiographic progression includes ground-glass opacifica-

tion and lobe thickening [17]. MERS-CoV (intact virus or

viral genome) is found at higher concentrations in the

lower respiratory tract than in the upper respiratory tract in

MERS patients and this may account for inefficient inter-

human transmission [15, 24]. Currently, no approved

therapeutics for patients with MERS or SARS are avail-

able, and clinical management has relied primarily on

supportive care.

Fig. 1 Maps of the severe acute respiratory syndrome (SARS) (a) and Middle East respiratory syndrome (MERS) (b) outbreaks with confirmed

case numbers

Therapeutics for MERS-CoV and SARS-CoV 1937

Page 4: Middle East Respiratory Syndrome and Severe Acute ... · The outbreaks of Middle East respiratory syndrome (MERS) and severe acute respiratory syndrome (SARS) were caused by emerging

2 Therapeutic Agents

2.1 Clinical Usage

2.1.1 Treatment of SARS

Effectiveness of antiviral treatments used during the SARS

epidemic has been mainly based on case studies and ret-

rospective analysis of patient cohorts. Few randomized,

blinded, clinical trials of anti-SARS treatments were per-

formed, which adds complexity when interpreting the

available data (Table 1). Ribavirin, a nucleoside analog

that prevents RNA and DNA virus replication, was initially

used in the treatment of SARS due to its broad-spectrum

efficacy. For example, in a Taiwanese study, 51 SARS

patients were treated daily with fluoroquinolone antibiotics,

[levofloxacin (500 mg) or moxifloxacin (400 mg)] fol-

lowing diagnosis. Out of 51 patients, 44 SARS patients

were also treated intravenously (IV) with 2000 mg of rib-

avirin then orally daily with 1200 mg while 7 SARS

patients did not receive ribavirin. Corticosteroids, IV

methylprednisolone, or oral prednisolone were adminis-

tered as needed to treat worsening lung infiltrates and fever

[25]. Ribavirin treatment led to hypoxia and anemia and

increased risk for death in SARS patients. In a retrospective

analysis, a cohort of 229 patients from Hong Kong, Sin-

gapore, and Toronto were treated with ribavirin in con-

junction with corticosteroids, immunoglobulins, and/or

antibiotics [26]; ribavirin did not demonstrate efficacy.

Patients in Hong Kong and Singapore were treated with

ribavirin at 1200 mg orally at diagnosis, followed by oral

treatments with 2400 mg daily, or continual IV ribavirin

therapy [8 mg/kg every 8 h (h)]. In Toronto, patients

received ribavirin IV treatment with 2000 mg, followed by

1000 mg every 6 h for 4 days, and 300 mg every 8 h for

3 days. Unfortunately, fatality rates were similar between

the ribavirin-treated and control groups. Later, researchers

demonstrated that the ribavirin dosage required to be

effective against SARS-CoV in vitro was not clinically

achievable [27]. Ribavirin treatment also resulted in

adverse effects including anemia, hypoxemia and

decreased hemoglobin levels, and did not improve patient

outcome [26]. Due to the increasing adverse effects and

lack of efficacy, Health Canada stopped permitting the use

of ribavirin for SARS [25].

Table 1 Drug regimens used in the treatment of SARS

Treatment plan Treatment outcome

Ribavirin (oral/IV)

Antibiotics

± corticosteroids

± immunoglobulin

No increased positive outcome with ribavirin

compared to controls [25, 26]

Increased risk of anemia, hypomagnesemia,

hypoxia, or bradycardia with ribavirin

compared to ribavirin-naive patients

[25, 246]

Ribavirin (oral/IV)

Lopinavir/ritonavir

± corticosteroids

Fatality or acute respiratory distress syndrome

(ARDS) was reduced significantly from 28.8

to 2.4% [27]

IFN-alfacon-1

± corticosteroids

± antibiotics

Increased oxygen saturation

Increased clearance of lung abnormalities

Slight increase in creatinine kinase

concentrations [29, 247]

Fluoroquinolone

(IV)

Azithromycin (IV)

IFN-a (IM)

± corticosteroids

± Immunoglobulins

± thymic peptides/

proteins

No increased positive outcome [248]

Quinolone (IV)

Azithromycin (IV)

± IFN-a

± corticosteroids

No increased positive outcome [248]

Levofloxacin

Azithromycin

± IFN-a

± corticosteroids

Increased survival

Increased clearance of lung abnormalities

[248]

IFN interferon, IM intramuscular, IV intravenous, SARS severe acute

respiratory syndrome

Fig. 2 Genomes of Middle East respiratory syndrome coronavirus

(MERS-CoV) and severe acute respiratory syndrome coronavirus

(SARS-CoV) indicating the open reading frames for nonstructural (1a

and 1b) and structural proteins (numbered 3–9, and E, M, N, S).

E envelope, M membrane, N nucleocapsid, S Spike

1938 J. Dyall et al.

Page 5: Middle East Respiratory Syndrome and Severe Acute ... · The outbreaks of Middle East respiratory syndrome (MERS) and severe acute respiratory syndrome (SARS) were caused by emerging

Additional studies tested the efficacy of ribavirin in

conjunction with lopinavir, an anti-retroviral agent. Lopi-

navir demonstrated in vitro activity against SARS-CoV

[28]. In a non-randomized, open-enrollment trial of 152

suspected SARS patients [27], all patients were treated

with ribavirin and corticosteroids similar to the previously

described studies. In addition, 41 of the confirmed SARS

patients were also treated with a combination of lopinavir

(400 mg) and ritonavir (100 mg). Mean viral loads in

nasopharyngeal swabs within this treatment group

decreased to undetectable levels by day 10. Overall, SARS-

related symptoms subsided, disease progression was

milder, and no adverse effects were reported as compared

to the historical control group.

In an open-label, non-randomized study of 22 SARS

patients, 9 patients who received subcutaneous (SC)

injections of interferon (IFN)a, alfacon-1, for 10 days at an

initial dose of 9 lg/day for 2 days increasing to 15 lg/daywith disease progression. All 9 patients survived with

minor adverse effects [29].

2.1.2 Treatment of MERS

The evaluation of treatments in MERS patients has been

hampered as high-quality clinical data from randomized

clinical trials are limited. Ribavirin (with or without IFN,

or corticosteroids) was the primary treatment during the

MERS outbreak. In a retrospective analysis, a cohort of 20

patients was treated with oral ribavirin and SC pegylated

IFN-a2a at a dose of 180 lg/week for 2 weeks (Table 2)

[30]. The initial dose of ribavirin was 2000 mg, followed

by a 200–1200 mg dose depending on creatinine clearance.

A group of 24 patients that received supportive care and

corticosteroids were considered the control group. At

14 days after confirmed diagnosis of MERS, survival was

increased in the treated group (70%) compared to the

control group (29%). By 28 days post-diagnosis, 30% of

treated subjects survived versus 17% of the control group

[30]. In an additional case study, a 69-year-old Greek

patient who contracted MERS in Jeddah was treated with

oral lopinavir/ritonavir (400/100 mg twice daily), pegy-

lated IFN (180 lg SC once per week for 12 weeks), and

ribavirin (2000 mg initial dose; 1200 mg every 8 h for

8 days, initiated on day 13 post-diagnosis). Two days after

treatment initiation, viremia could not be detected; how-

ever, viral RNA was detected in several patient samples

(feces, respiratory secretions, and serum) up to 14 weeks

post-diagnosis. Despite prolonged survival, the patient

succumbed from septic shock 2 months post-diagnosis

[31]. An ongoing randomized clinical trial in Saudi Arabia

is evaluating treatment of MERS patients with IFN-b1b in

combination with lopinavir/ritonavir [32].

2.2 Drugs with Repurposing Potential

for Treatment of Coronaviral Infections

Drug repurposing is an attractive alternative drug discovery

strategy because it eliminates many steps usually required

at the early phase of drug development. Over the past

decade, interest in drug repurposing has increased as

pharmaceutical companies are challenged with decreasing

product pipelines, high costs associated with de novo drug

discovery, and the imminent expiration of many drug

patents. Some examples for successfully repurposed drugs

include Viagra (Pfizer) for erectile dysfunction (original

indication: angina) and raloxifene (Eli Lilly) for treatment

of invasive breast cancer (original indication:

osteoporosis).

The time required for traditional drug development is

often discordant with the urgent need for novel therapies

for emerging infectious diseases such as SARS and MERS.

Outbreaks can occur anywhere in the world and frequently

in resource-limited settings. Commonly, the treatment

strategies that are available for emerging infectious dis-

eases are less than adequate to improve patient outcome.

Although specific antivirals for MERS-CoV and SARS-

CoV are in development, drug repurposing could present

an important arm in generating additional therapeutics for

future coronaviruses. First, if these drugs are confirmed to

have beneficial effects in vitro and in animal studies, they

could be used to build a panel of approved drugs for use as

a first-line of defense for newly emerging coronaviruses.

Second, these drugs could be made accessible relatively

quickly to patients under Emergency Use Authorization.

Extending the choices of treatment by generating a panel of

Table 2 Drug regimens used in the treatment of MERS

Treatment plan Treatment outcome

Ribavirin (oral/

IV)

IFN-a2b

Corticosteroids

Late treatment administration. Disease

progression delayed—all patients died [249]

Ribavirin (oral/

IV)

PEGylated IFN-

a2a (IV)

± corticosteroids

Treatment initiated 0–8 days after diagnosis

Adverse effects: significant decreases in

hemoglobin and absolute neutrophil count

(baseline count lower in treatment group) [30]

Ribavirin (oral/

IV)

Lopinavir/

ritonavir

IFN-a2b

No detectable viral RNA in serum after 2 days of

therapy

Adverse effects: ribavirin discontinued due to

jaundice, hyperbilirubinemia

Died of septic shock 2 months, 19 days after

diagnosis [31]

IFN interferon, IV intravenous, MERS Middle East respiratory

syndrome

Therapeutics for MERS-CoV and SARS-CoV 1939

Page 6: Middle East Respiratory Syndrome and Severe Acute ... · The outbreaks of Middle East respiratory syndrome (MERS) and severe acute respiratory syndrome (SARS) were caused by emerging

broad-spectrum antivirals would provide a real improve-

ment to healthcare communities struggling to cope during

an outbreak of emerging infections. A great example of

how repurposing can benefit in the search of treatments for

emerging infections is the drug zidovudine. Zidovudine

was originally developed in 1964 as a cancer drug. In 1985,

zidovudine was found to be active against human

immunodeficiency virus (HIV), and 2 years later it became

the first drug to be approved for the treatment of acquired

immunodeficiency syndrome (AIDS) [33].

A number of research groups have identified and

investigated the usefulness of approved drugs for the

treatment of viral infections including coronaviruses.

Below, we summarize several drug classes with antiviral

activity against MERS-CoV and SARS-CoV that have

repurposing potential (Fig. 3, Table 3). Some of the drugs

described have activity against other virus families indi-

cating potential broad-spectrum applications and have

already been in clinical use for treating other viral infec-

tions. We would like to emphasize that none of the thera-

peutics described in this section are recommended for

clinical use outside a clinical trial setting.

2.2.1 Antidiarrheal Agents

Loperamide, an approved anti-diarrheal agent, is on the

World Health Organization (WHO) Model List of Essential

Medicines and is available in many countries. The drug

acts on the opioid receptor and reduces intestinal motility

[34]. Results from pharmacokinetic (PK) studies show that

oral loperamide is well absorbed from the gut with less

than 1% of the drug entering systemic circulation [35].

Loperamide demonstrated anti-MERS-CoV, anti-SARS,

and anti-HCoV229E activity in an in vitro screen of

approved drugs [36], although the mechanism of action is

unknown. Interestingly, loperamide was suggested for

limiting gastrointestinal fluid and electrolyte losses in

patients with Ebola virus disease (EVD) [37].

2.2.2 Antimalarial Agents

The antimalarial agents, chloroquine (CQ), amodiaquine,

and mefloquine have activity against SARS-CoV and

MERS-CoV in vitro [36, 38, 39]. CQ is a U.S. Food and

Drug Administration (FDA)-approved antimalarial agent

that is also used to treat autoimmune disease such as

rheumatoid arthritis due to its anti-inflammatory effects

[40]. CQ has activity against a number of viruses in vitro

and in vivo including flaviviruses [dengue virus (DENV)],

Togaviruses [chikungunya virus (CHIKV)], paramyx-

oviruses (Hendra, Nipah virus), influenza viruses, HIV,

and filoviruses [Ebola virus (EBOV)] [41–47].

Several mechanisms of action have been identified for

the antiviral effect of CQ and suggest that the drug acts

nonspecifically at virus entry or at the later stages of virus

production. CQ accumulates within acidic organelles such

as endosomes, Golgi vesicles, and lysosomes, where the

drug is protonated resulting in increased pH within the

vesicle [48]. Viruses depend on these acidic organelles for

entry, viral replication, and maturation of virus progeny.

Similarly, MERS-CoV entry into cells depends on several

proteases. Dipeptidyl peptidase 4 (DPP4) acts as functional

virus receptor [49], and cellular proteases [e.g. type II

transmembrane serine protease (TMPRSS2) and members

of the cathepsin family] activate the viral spike (S) glyco-

protein [50]. CQ may have an effect on any of these pro-

teases. CQ also affects the glycosylation step within the

Golgi that directs trafficking and maturation of viral pro-

teins [51–53]. For SARS-CoV, the antiviral activity of CQ

has also been attributed to a deficit in glycosylation of the

receptor angiotensin-converting enzyme 2 (ACE2) [54].

The broad-spectrum antiviral activity makes CQ an

attractive antiviral for repurposing and treating coronaviral

and other emerging viral infections. In vivo activity of CQ

in MERS or SARS animal models has not yet been

reported. However, the antiviral activity of the drug has

been evaluated against other viruses in preclinical and

clinical studies with mixed results. CQ plasma steady state

concentrations in mice are similar to those reported for

humans (10 lM) and are within range of the EC50 values

determined for MERS-CoV (3.6 lM) and SARS-CoV

(2.3 lM) [43, 55]. Preclinical studies with CQ in mice

against other viruses have shown survival benefits for

influenza and EBOV infections.

In clinical studies, CQ was effective at reducing viral

loads in asymptomatic HIV patients [56, 57], but results of

CQ treatment of CHIKV and DENV infections were mixed

[58, 59].

In summary, CQ has broad-spectrum potential and the

information gained from studies on other viruses can be

used to plan the most appropriate strategies for evaluating

its specific clinical value for treating for MERS-CoV and

SARS-CoV infections. CQ has several advantages includ-

ing rapid absorption from gastrointestinal tract, low cost,

cFig. 3 Candidate drugs for repurposing for coronaviral infections.

Several drug classes (A through I) have been studied, and the steps/

processes of the viral replication cycle that they most likely target are

indicated. AKT serine/threonine kinase, CAD cationic amphiphilic

drug, Cyps cytochrome P-450s, E envelope, ER endoplasmic retic-

ulum, ERGIC ER–Golgi intermediate compartment, ERK extracellu-

lar signal-reduction kinase, IFN interferon, MAPK mitogen-activated

protein kinase, M membrane, MPA mycophenolic acid, mTOR

mechanistic target of rapamycin, N nucleocapsid, NFAT nuclear

factor of activated T cells, ORF open reading frame, PI3K phospho-

inositide 3-kinase, S Spike

1940 J. Dyall et al.

Page 7: Middle East Respiratory Syndrome and Severe Acute ... · The outbreaks of Middle East respiratory syndrome (MERS) and severe acute respiratory syndrome (SARS) were caused by emerging

Therapeutics for MERS-CoV and SARS-CoV 1941

Page 8: Middle East Respiratory Syndrome and Severe Acute ... · The outbreaks of Middle East respiratory syndrome (MERS) and severe acute respiratory syndrome (SARS) were caused by emerging

and very effective biodistribution. CQ may be an excellent

candidate for combinatorial treatments with other antivi-

rals. However, considerable challenges remain for the

treatment of viral infections including increased under-

standing of the pharmacodynamics of CQ, achievement of

sufficient plasma concentrations in patients, and toxicity

concerns [60]. Importantly, hydroxychloroquine, a CQ

derivate, may provide an alternative due to lower toxicity

and similar pharmacology profile [55].

A related antimalarial drug, amodiaquine, also has

activity in vitro against MERS-CoV and SARS-CoV [38].

Previous investigations have demonstrated that amodi-

aquine inhibits filovirus replication, and the mechanism of

action is hypothesized to be similar to that of CQ [43].

Amodiaquine is well tolerated and is commonly used for

malaria treatments in many countries. Further, amodi-

aquine in combination with artesunate was administered to

EVD patients during the 2013–2016 epidemic, and the

resulting decrease in fatality rates may have been associ-

ated with the use of amodiaquine as an antimalarial agent

[61]. Nonhuman primate (NHP) studies are currently

underway to investigate the effect of amodiaquine treat-

ment on EVD [62].

Mefloquine, a synthetic analog of quinine, is another

antimalarial drug with activity against MERS-CoV and

SARS-CoV [38]. It belongs to the WHO Model List of

Essential Medicines. Mefloquine is known to penetrate the

blood-brain barrier and was found to inhibit JC virus

infection and replication at concentrations generally

achieved in the brains of patients given mefloquine for

malaria [63] leading to the clinical evaluation of this drug

for the treatment of progressive multifocal leukoen-

cephalopathy [64, 65]. In 2013, the FDA added a boxed

warning to the US label of mefloquine regarding the

potential for neuropsychiatric side effects. Additional

investigations are warranted to determine if amodiaquine

or mefloquine have value for repurposing for treatment of

MERS or SARS.

2.2.3 Cyclophilin Inhibitors

Cyclophilins are ubiquitous host proteins believed to have

multiple roles in trafficking, protein folding and T cell

activation [66]. Cyclosporine A (CysA), forms a complex

with cyclophilin A, thereby blocking T cell activation.

CysA is licensed for use in organ transplantation to sup-

press the immune response. CysA has also been shown to

inhibit coronaviruses including SARS-CoV and MERS-

CoV effectively in cell culture [67, 68]; however, the

mechanism has yet to be determined. There is increasing

evidence that cyclophilins are involved in viral replication

of RNA viruses such as hepatitis C virus (HCV) and West

Nile virus, and this may also apply to coronaviruses [69].

Although the immunosuppressive properties of CysA are

considered a risk for treating viral infections in patients,

nonimmunosuppressive analogs of CysA that bind to

cyclophilins with higher affinity have been developed and

some are in clinical trials as HCV therapeutics [70–72].

2.2.4 Interferons

Interferons (IFNs) are approved by the FDA for other

indications such as hepatitis C. Although IFN-a reduced

SARS-CoV replication in mice and NHPs [73, 74], efficacy

of IFN-a treatment in SARS patients was mixed (see

Sect. 2.1.1). From in vitro studies, another type I inter-

feron, IFN-b1a, may be more effective than IFN-a either

alone or in combination with IFN-c [75–77]. Combinations

of IFN-b and -c were synergistic against SARS-CoV

in vitro [77].

With regards to MERS, in vitro and in vivo preclinical

studies have indicated that IFN-a2b alone or in combina-

tion with ribavirin, may have a therapeutic effect if given

early in disease [78, 79]. In clinical trials, however, IFN-

a2b (given in combination with other treatments) did not

lead to a significant benefit to patients (see Sect. 2.1.2).

IFN-b1a (EC50 = 1.37 IU/mL) was superior in activity

against MERS-CoV infection in vitro compared to IFN-

a2a, IFN-a2b, and IFN-c; these IFNs had EC50 values of

160.8, 21.4, and 56.5 IU/mL, respectively [80]. IFN-b1b is

currently under evaluation for MERS-CoV in a randomized

clinical trial (in combination with lopinavir/ritonavir) [32].

Investigating the IFN-b subtypes (1a and 1b) in combina-

tion with other antivirals may be worthwhile as potential

synergistic combinations could reduce the effective drug

dosage and IFN-associated adverse effects.

2.2.5 Kinase Inhibitors

Many cellular processes are regulated independently of

changes in transcription or translation through kinase-me-

diated cell signaling pathways. As a testament to the bio-

logical importance of kinases, there have been over 500

kinases identified along with more than 900 genes encod-

ing proteins with kinase activity [81, 82]. As of April 2015,

28 kinase inhibitors have been granted approval by the US

FDA with over half gaining approval from 2012–2015.

Further, kinases are the most frequently targeted gene class

in cancer therapy, second only to the G protein-coupled

receptors as therapeutic targets [83, 84].

The therapeutic potential for host-targeted

immunomodulatory agents in viral infections has received

considerable attention [85–87]. Recently, Dyall et al.

identified two Abelson (Abl) kinase inhibitors (imatinib

and dasatinib) that inhibited MERS-CoV and SARS-CoV

infection through a cell-screening assay [38]. Both

1942 J. Dyall et al.

Page 9: Middle East Respiratory Syndrome and Severe Acute ... · The outbreaks of Middle East respiratory syndrome (MERS) and severe acute respiratory syndrome (SARS) were caused by emerging

Table

3Clinically

developed

drugswithactivityagainst

MERS-CoV

andSARS-CoV

Drugclassmem

bers

Activityagainst

coronavirusesa

Activityagainstother

viruses

Clinical

status

Knownsafety

issues

Comments

Antidiarrhealagents

Loperam

ide

hydrochloride

MERS-CoV:

4.8

lM[36]

SARS-CoV:

5.9

lM

HCoV-229E-G

FP:

4lM

[36]

Approved

fortreatm

entofdiarrhea

Welltolerated,

commonly

used

WHO

list

ofessential

medicines

Antimalaria

agents

Amodiaquine

hydrochloride

MERS-CoV:

6.2

lM[38]

SARS-CoV:

1.3

lM[38]

EBOV

[43];MARV

[43];DENV

[250]

Approved

fortreatm

entofmalaria

Welltolerated,

commonly

used

Chloroquine

diphosphate

MERS-CoV:

3–6.3

lM

[36,38]

SARS-CoV:

4.1–8.8

lM

[36,38,39]

Invitro:

CHIK

V[251,252];DENV

[42,47];

HeV

,NiV

[46];HIV

-1[41];

FLUAV

[44];EBOV,MARV

[43];

SFV,SIN

V[45]

Invivo:

Mixed

dataonefficacy

against

EBOV

inmice[43,60];noefficacy

against

EBOV

inham

sters,guinea

pigs[60];FLUAV:70%

survivalin

mice[253]

Approved

fortreatm

entofmalaria

Clinical

trials

fortreatm

entofCHIK

V,

DENV,FLUAV,andFLUBV

infectionsshowed

noim

pacton

disease

[58,59,254]

Welltolerated,

commonly

use

WHO

list

ofessential

medicines;

WHO

listofpotential

EVD

treatm

ents;dosing,form

ulation

needto

beoptimized

fortreatm

ent

ofviral

infections[54]

Hydroxychloroquine

sulfate

MERS-CoV:

8.3

lM[38]

SARS-CoV:

8.0

lM[38]

Invitro:

DENV

[42];HIV

-1[255];JC

V[63]

Approved

fortreatm

entofmalaria

Welltolerated,

commonly

used

WHO

list

ofessential

medicines

Mefloquine

MERS-CoV:

7.4

lM[38]

SARS-CoV:

15.6

lM[38]

Approved

fortreatm

entofmalaria

Clinical

trials

fortreatm

entofJC

V

infection:mixed

results[64,65]

Black

boxwarning:

potential

neuropsychiatric

sideeffects

WHO

list

ofessential

medicines

Cyclophilin

inhibitors

CyclosporinA

MERS-CoV

[67]

SARS-CoV

[68]

HCV

[256];WNV

[257];JEV

[258]

VSV

[259];HIV

-1[260]

Approved

forim

munosuppression

duringorgan

transplantation

Immunosuppression

undesirable

for

infectiousdiseases

WHOlistofessentialmedicines;non-

immunosuppressiveanalogsare

available

[70–72]

Interferons

IFN-a2a

MERS-CoV:

160.8

U/m

L[80]

Approved

fortreatm

entofhepatitis

B

andC

Welltolerated

Therapeutics for MERS-CoV and SARS-CoV 1943

Page 10: Middle East Respiratory Syndrome and Severe Acute ... · The outbreaks of Middle East respiratory syndrome (MERS) and severe acute respiratory syndrome (SARS) were caused by emerging

Table

3continued

Drugclassmem

bers

Activityagainst

coronavirusesa

Activityagainstother

viruses

Clinical

status

Knownsafety

issues

Comments

IFN-a2b

MERS-CoV:

21.4

U/m

L[80]

SARS-CoV:6500-

4950U/m

L[75]

Approved

fortreatm

entofmelanoma

IFN-b1a,

1b

MERS-CoV:

1.4

U/m

L[80]

SARS-CoV:

95–105U/m

L

[75]

Approved

fortreatm

entofmultiple

sclerosis

IFN-c

MERS-CoV:

56.5

U/m

L[80]

SARS-CoV:

1700–2500U/

mL[115]

Approved

fortreatm

entofchronic

granulomatousdisease

Kinaseinhibitors

Dabrafenib

MERS-CoV:45%

inhibitionat

10lM

[89]

Approved

fortreatm

entofcancers

Welltolerated

Dasatinib

MERS-CoV:

5.5

lM[38]

SARS-CoV:

2.1

lM[38]

Invitro:

BKPyV

[261];EBOV

[100];HIV

-1

[262];DENV

[263]

Invivo:

VACV:noactivityin

mice[264]

Approved

fortreatm

entofcancers

Welltolerated

ABL1inhibitor,CAD

Immunosuppressiveeffectsin

vivo

may

precludeuse

asanti-infective

Everolimus

MERS-CoV:56%

inhibitionat

10lM

[89]

DENV,CPXV,RSV,FLUAV

[265]

Approved

immunosuppressantfor

cancertreatm

entandpreventionof

organ

rejection;reducedincidence

of

HHV-5

incardiacandrenaltransplant

patients

[266]

Welltolerated

mTOR

inhibitor[267]

Imatinib

mesylate

MERS-CoV:

17.7

lM[38]

SARS-CoV:

9.8

l M[38]

Invitro:

BKPyV

[261];HHV

[268];HCV

[149];MPXV,VACV,VARV[264]

Invivo:

VACV:100%

survival

inmice[264]

Approved

fortreatm

entofcancers

Welltolerated

ABL1inhibitor

Miltefosine

MERS-CoV:28%

at10lM

[89]

HSV-2

[269]

Antimicrobialapproved

fortreatm

entof

leishmaniasis;investigational

drugfor

treatm

entofam

oebainfections

Welltolerated

Aktinhibitor

WHO

list

ofessential

medicines

1944 J. Dyall et al.

Page 11: Middle East Respiratory Syndrome and Severe Acute ... · The outbreaks of Middle East respiratory syndrome (MERS) and severe acute respiratory syndrome (SARS) were caused by emerging

Table

3continued

Drugclassmem

bers

Activityagainst

coronavirusesa

Activityagainstother

viruses

Clinical

status

Knownsafety

issues

Comments

Nilotinib

MERS-CoV:

5.5

lM[38]

SARS-CoV:

2.1

lM[38]

Selumetinib

sulfate

MERS-CoV:

C95%

inhibition

at10lM

[89]

InPhaseII/IIItrialsforcancertreatm

ent

Welltolerated

MEK

inhibitor

Sirolimus

MERS-CoV:61%

inhibitionat

10lM

[89]

HIV

-1[270];HHV-5

[271]

Approved

immunosuppressantin

transplant;reducedviral

replicationin

HIV

-orHCV-positivetransplant

patients

[272–274]

Welltolerated

mTOR

inhibitor

Sorafenib

MERS-CoV:93%

inhibitionat

10lM

[89]

BKPyV

[261];HHV

[275];EV71

[276];RVFV

[277]

Approved

fortreatm

entofcancers

Welltolerated

Trametinib

MERS-CoV:

C95%

inhibition

at0.1

lM[89]

Approved

fortreatm

entofcancers

Welltolerated

Wortmannin

MERS-CoV:40%

inhibitionat

10lM

[89]

HSV-2

[269];HIV

-1[278]

Indevelopmentfortreatm

entofcancer

Toxicityissues.

Derivatives

arein

PhaseIclinical

trials

Neurotransm

itterinhibitors

Astem

izole

MERS-CoV:

4.9

lM[38]

SARS-CoV:

5.6

lM[38]

Previouslyapproved

antihistamine

Withdrawnin

1999

because

ofrare

arrhythmias

Benztropine

mesylate

MERS-CoV:

16.6

lM[38]

SARS-CoV:

21.6

lM[38]

HCV

[102],EBOV,MARV

[100,101,104]

Approved

anticholinergic

fortreatm

ent

ofParkinson’s

disease

Welltolerated

Chlorphenoxam

ine

MERS-CoV:

12.7

lM[38]

SARS-CoV:

20.0

lM[38]

Approved

antihistamineand

anticholinergic

fortreatm

entof

Parkinson’s

Disease

Welltolerated

Chlorpromazine

hydrochloride

MERS-CoV:

4.9–9.5

lM

[36,38]

SARS-CoV:

8.8–13.0

lM

[36,38]

EBOV

[96];JU

NV

[94];SV40[279]

Approved

fortreatm

entof

schizophrenia

Welltolerated

CAD

Therapeutics for MERS-CoV and SARS-CoV 1945

Page 12: Middle East Respiratory Syndrome and Severe Acute ... · The outbreaks of Middle East respiratory syndrome (MERS) and severe acute respiratory syndrome (SARS) were caused by emerging

Table

3continued

Drugclassmem

bers

Activityagainst

coronavirusesa

Activityagainstother

viruses

Clinical

status

Knownsafety

issues

Comments

Clomipramine

hydrochloride

MERS-CoV:

9.3

lM[38];

SARS-CoV:

13.2

lM[38]

EBOV

[100]

Approved

fortreatm

entofdepression

Welltolerated

WHO

list

ofessential

medicines

Fluphenazine

hydrochloride

MERS-CoV:

5.9

lM[38];

SARS-CoV:

21.4

lM[38]

HCV

[149];EBOV

[100]

Approved

fortreatm

entofchronic

psychoses

Welltolerated

WHO

list

ofessential

medicines;

CAD

Fluspirilene

MERS-CoV:

7.5

lM[38]

SARS-CoV:

5.9

lM[38]

Approved

fortreatm

entof

schizophrenia

Welltolerated

Promethazine

hydrochloride

MERS-CoV:

11.8

lM[38]

SARS-CoV:

7.5

lM[38]

Antipsychoticapproved

forsedation

Welltolerated

Thiothixene

MERS-CoV:

9.3

lM[38]

SARS-CoV:

5.3

lM[38]

Antipsychoticapproved

fortreatm

entof

schizophrenia

Welltolerated

Triethylperazine

maleate

MERS-CoV:

7.8

lM[38]

SFV

[45],CHIK

V[45,99]

Approved

antiem

etic

Welltolerated

Triflupromazine

hydrochloride

MERS-CoV:

5.8

lM[38];

SARS-CoV:

6.4

lM[38]

Approved

antipsychotic

Serioussideeffects

includeakathisia.

CAD

Nucleicacid

synthesis

inhibitors

Gem

citabine

hydrochloride

MERS-CoV:

1.2

lM[38]

Invitro:

FLUAV

[125]

Invivo:

MiceMuLV

[124]

Approved

fortreatm

entofcancers

Welltolerated

WHO

list

ofessential

medicines

Mizoribine

SARS-CoV:3.5-

16lg

/mL[114]

or[

40lM

[111]

HCV

[123];BVDV

[280]

Approved

immunosuppressantin

organ

transplantationandrheumatic

diseases.

Welltolerated

Mycophenolicacid

MERS-CoV:

0.17lg

/mL

[112]or2.87lM

[80]SARS-CoV:

[30lM

[111]

DENV

[118];CHIK

V[252];FLUAV

[112];SFV,SIN

V[45]

Approved

immunosuppressantin

organ

transplantation

FDA

alert:risk

of

activationoflatent

herpes

infections

1946 J. Dyall et al.

Page 13: Middle East Respiratory Syndrome and Severe Acute ... · The outbreaks of Middle East respiratory syndrome (MERS) and severe acute respiratory syndrome (SARS) were caused by emerging

Table

3continued

Drugclassmem

bers

Activityagainst

coronavirusesa

Activityagainstother

viruses

Clinical

status

Knownsafety

issues

Comments

Proteaseinhibitors

Lopinavir

Invitro:

MERS-CoV:

8lM

[36]

SARS-CoV:

24.4

lM[36]

Invivo:

67%

survival

in

MERS-infected

NHPs[121]

HIV

-1[136]

HPV

[281]

Approved

fortreatm

entofHIV

infections

Clinical

trialfortopical

treatm

entof

cervical

cancer

Clinical

trialin

SARSpatients

[27]

Welltolerated

Lopinavir/ritonavir:WHO

listof

essential

medicines

E-64-D

MERS-CoV:

1.27lM

[38];

SARS-CoV:

0.76lM

[38]

EBOV

[134,135]

Cysteineproteaseinhibitor

PhaseIIIfortreatm

entofmuscular

dystrophy

Welltolerated

K1177

MERS-CoV,

SARS-CoV,

HCoV-229E

(VLP)[133]

EBOV,MARV,NiV

(VLP)[133]

Cysteineproteaseinhibitor

Inclinical

developmentfortreatm

entof

Chagas

disease

Welltolerated

Cam

ostat

mesylate

Invitro:

MERS-CoV

[132],SARS-

CoV

[132]

Invivo:

SARS:activityin

mice[133]

Invitro:

FLUAV,FLUBV

[130];no

inhibitionofEBOV

[133]

Cellularserineproteaseinhibitor

Inclinical

developmentforchronic

pancreatitis

Welltolerated

SARS-CoVspread

isdriven

byserine

proteaserather

than

cysteine

protease[133]

Protein

synthesis

inhibitors

Anisomycin

MERS-CoV:

0.003lM

[38]

SARS-CoV:

0.19lM

[38]

PV

[147];EMCV

[146]

Antibioticin

clinical

developmentfor

treatm

entofam

oebiasis

Emetine

hydrochloride

MERS-CoV:

0.014lM

[38]

SARS-CoV:

0.05lM

[38]

FLUAV

[112];EMCV

[146]

Approved

antibioticfortreatm

entof

amoebiasisin

somecountries

Sideeffectsinclude

nausea;

derivatives

withless

side

effectsavailable

Omacetaxine

mepesuccinate

MERS-CoV:

9.07lM

[38]

MHV:0.012lM

[139]

HBV

[148]

Approved

forchronicmyeloid

leukem

iaWelltolerated

Therapeutics for MERS-CoV and SARS-CoV 1947

Page 14: Middle East Respiratory Syndrome and Severe Acute ... · The outbreaks of Middle East respiratory syndrome (MERS) and severe acute respiratory syndrome (SARS) were caused by emerging

Table

3continued

Drugclassmem

bers

Activityagainst

coronavirusesa

Activityagainstother

viruses

Clinical

status

Knownsafety

issues

Comments

Selectiveestrogen

response

modulator

Tam

oxifen

citrate

MERS-CoV:

10.1

lM[38];

SARS-CoV:

92.9

lM[38]

HCV

[282];HSV-1

[283]

Approved

fortreatm

entofbreastcancer

Black

boxwarning:

uterinecancer,

bloodclots,stroke

WHO

list

ofessential

medicines;

CAD

Toremifenecitrate

MERS-CoV:

12.9

lM;SARS-

CoV:12lM

[38]

Invitro:

HCV

[149];EBOV

[100,151];

SUDV;RAVV;MARV

[151]

Invivo:

EBOV:50%

survival

inmice[151]

Approved

fortreatm

entofbreast

cancer.

Black

boxwarning:

cardiaceffect

(QT

prolongation)in

patientswith

hypokalem

ia

WHO

list

ofpotential

EVD

treatm

ents;CAD

Sterolmetabolism

inhibitors

Terconazole

MERS-CoV:

12.2

lM[38];

SARS-CoV:

15.3

lM[38]

EBOV-V

LP[152];EVD

[100]

Approved

topical

antifungal

for

treatm

entofvaginal

yeast

infections

Would

requireIN

D

fororaluse

CAD

Triparanol

MERS-CoV

5.3

lM[38]

EBOV-V

LP[152]

Developed

forloweringserum

cholesterol;withdrawn

Acute

cataract

form

ation

CAD

ABL1abelsonmurineleukem

iaviral

oncogenehomolog1,Akt

protein

kinaseB,BKPyV

BK

polyomavirus,

BVDV

Bovineviral

diarrhea

virus,

CAD

cationic

amphiphilic

drug,CHIK

Chikungunyavirus,

CPXV

Cowpox

virus,

CVsCoxsackie

viruses,

DENV-1

Denguevirus-1,DENV-2

Denguevirus-2,EBOV

Ebola

virus,

EC50effectiveconcentration

50,EMCV

Encephalomyocarditisvirus,EV71Enterovirus71,FCoVFelinecoronavirus,FDAFoodandDrugAdministration,FLUAVInfluenza

Avirus,FLUBVInfluenza

Bvirus,HBVHepatitisBvirus,

HCV

hepatitis

Cvirus,

HeV

Hendra

virus,

HHV-1

Human

herpesvirus-1(herpes

simplexvirus-1),

HHV-2

human

herpresvirus-2(herpes

simplexvirus-2),

HHV-5

Human

herpesvirus-5

(cytomegalovirus),HIV-1

Human

immunodeficiency

virus-1,HIV-2

Human

immunodeficiency

virus-2,HPVHuman

papillomavirus,HRVsHuman

rhinoviruses,INDInvestigationalnew

drug,

JCVJohnCunningham

virus,JU

NVJunin

virus,MARVMarburg

virus,MEK

MAPK/ERK

kinase,

MERS-CoVMiddle

Eastrespiratory

syndromecoronavirus,MHVMouse

hepatitis

virus,

MPXVMonkeypoxvirus,mTORmechanistictargetofrapam

ycin,MuLVMurineleukem

iavirus,NiV

Nipah

virus,PVPoliovirus,RAVVRavnvirus;RSVRespiratory

syncytialvirus,RVFVRift

Valleyfever

virus,SARS-CoVSevereacuterespiratory

syndromecoronavirus,SFVSem

likiforestvirus,SFTSVSeverefever

withthrombocytopeniasyndromevirus,SINVSindbisvirus,SUDV

Sudan

virus,SV40Sim

ianvirus40;VACVVaccinia

virus,VARVVariola

virus,VLPvirus-likeparticles,WHO

WorldHealthOrganization

aAntiviral

activityisexpressed

interm

sofEC50unless

otherwisenoted

1948 J. Dyall et al.

Page 15: Middle East Respiratory Syndrome and Severe Acute ... · The outbreaks of Middle East respiratory syndrome (MERS) and severe acute respiratory syndrome (SARS) were caused by emerging

compounds significantly inhibited MERS-CoV and SARS-

CoV with micromolar EC50 values and low cytotoxicity.

Abl2 has been identified as critical for MERS-CoV and

SARS-CoV virus entry, and may be the target that imatinib

inhibits to block entry of both viruses [88]. A recent sys-

tems kinome analysis investigation of in vitro MERS-CoV

infection suggested that ERK/MAPK and PI3 K/Akt/

mTOR signaling pathways were specifically modulated

during infection [89]. Subsequent analysis of licensed

kinase inhibitors targeting these pathways demonstrated

that kinase inhibitors targeting the ERK/MAPK signal

pathway (selumetinib and trametinib) inhibited MERS-

CoV infection by C 95% when added pre- or post-infection

[89]. Further, trametinib demonstrated significantly stron-

ger inhibitory activity against MERS-CoV than selume-

tinib suggesting that specific intermediates of the ERK/

MAPK signaling pathway may represent crucial foci dur-

ing early (viral entry) and late (viral replication) events in

the viral life cycle. In contrast, sorafenib, an inhibitor of

Raf-1 and B-Raf, components of the ERK/MAPK signaling

pathway, and vascular endothelial growth factor receptor 2

(VEGFR2), inhibited MERS-CoV infection by [90%

when added to cells prior to infection; however, the inhi-

bitory activity was reduced to \30% when added post-

infection suggesting Raf kinases were primarily involved

in early viral life cycle events. In addition, the inhibitory

activity dabrafenib, a Raf kinase inhibitor, was also largely

ablated when added post-infection. Miltefosine, an alkyl

phospholipid, considered to be an inhibitor of protein

kinase B (Akt), garnered FDA approval for infectious

disease-related treatments (cutaneous or mucosal leishma-

niasis) [90]. In 2013, miltefosine became directly avail-

able from the US Centers for Diseases Control and

Prevention for the treatment of free-living amoeba infec-

tions [91]. Pre-treatment of cells with miltefosine reduced

MERS-CoV infection by 28%, but had no effect when

added post-infection [89]. In contrast, inhibition of mTOR

with sirolimus or everolimus reduced MERS-CoV infec-

tion by * 60% when added prior pre- or post-infection

suggesting a critical role for mTOR in MERS-CoV infec-

tion. A recent clinical investigation by Wang et al. [92]

evaluated sirolimus and corticosteroids in addition to

standard antiviral treatment in a randomized controlled trial

in patients with severe H1N1 pneumonia and acute respi-

ratory failure [92]. Importantly, the addition of sirolimus

was associated with improved patient outcomes including

decreased hypoxia and multi-organ dysfunction, reduced

mean times for liberation from mechanical ventilation, and

increased clearance of virus. Thus, it may be prudent to

extend the study of repurposed kinase inhibitors beyond

stand-alone therapeutic investigations and also consider

their potential as adjunctive therapies.

2.2.6 Neurotransmitter Inhibitors

Numerous neurotransmitter receptors inhibitors showed

activity against MERS-CoV and SARS-CoV infection [38].

These drugs were initially developed as antipsychotics,

antihistamines, and sedatives. Five neurotransmitter

receptor antagonists belong to the chemical class of phe-

nothiazines: chlorpromazine, triflupromazine, thiethylper-

azine, promethazine and fluphenazine. Phenothiazines were

breakthrough medications developed in the 1950s for

treating mental health patients and reduced episodes of

bizarre behavior, hallucinations, and irrational thoughts

[93]. Although phenothiazines primarily block dopamine

receptors, they also have anticholinergic, antihistamine,

and antiemetic effects.

The phenothiazines, chlorpromazine and triflupro-

mazine, are approved antipsychotics. Chlorpromazine has

been used off-label for short-term treatment of nausea and

migraines. Triflupromazine is used to treat severe emesis,

but the drug has more serious side effects than chlorpro-

mazine including akathisia and tardive dyskinesia. The

antiviral effect of chlorpromazine has been extensively

studied, and the drug interferes with clathrin-mediated

endocytosis, a process that many viruses exploit for host

cell entry. Chlorpromazine inhibits entry of Junin virus

[94], West Nile virus [95], EBOV [96], HCV [97], and

Japanese encephalitis virus [98] suggesting broad-spectrum

activity that could be exploited early during a novel virus

outbreak. Chlorpromazine may have similar effects on

coronaviruses as the drug effectively inhibits MERS-CoV,

SARS-CoV, and human coronavirus 229E expressing

green fluorescent protein [36, 38]. However, time-of-ad-

dition studies indicate that the inhibitory activity against

MERS-CoV is retained whether added pre- or post-infec-

tion suggesting that there are additional effects to clathrin-

mediated entry impairment [36].

Thiethylperazine is an approved antiemetic. Both

chlorpromazine and thiethylperazine have been shown to

inhibit alphaviruses, Semliki forest virus (SFV) and

chikungunya virus (CHIKV) [45, 99]. As these drugs cross

the brain-blood barrier, use of these drugs could be bene-

ficial in the treatment of CHIKV, including common neu-

rologic complications. Promethazine is an antihistamine

used as a sedative in many countries under different brand

names, but also acts as a weak anti-psychotic activity.

Fluphenazine is a common antipsychotic used to treat

chronic psychoses (primarily schizophrenia) and belongs to

the WHOModel List of Essential Medicines. Promethazine

and fluphenazine have shown in vitro activity against

MERS-CoV and SARS-CoV and may have value as can-

didates for repurposing for coronaviral infections [38].

Benztropine mesylate, an approved anticholinergic used

to treat Parkinson’s, had activity against MERS-CoV and

Therapeutics for MERS-CoV and SARS-CoV 1949

Page 16: Middle East Respiratory Syndrome and Severe Acute ... · The outbreaks of Middle East respiratory syndrome (MERS) and severe acute respiratory syndrome (SARS) were caused by emerging

SARS-CoV [38]. Benztropine was also identified in other

screens of clinically approved drugs for antiviral activities

against HCV and EBOV [100–102]. Although the detailed

mechanism of action is unknown, HCV studies indicate

that benztropine inhibits at a virus entry step, while not

interfering with viral genome replication, transcription or

production of viral progeny or virus production of viral

progeny [102]. It has been argued that a virus entry inhi-

bitor may have value in decreasing the incidence of relapse

in chronic HCV patients that receive liver transplants

[102]. However, the peak plasma concentrations of ben-

ztropine may be too low to be effective for treating an

acute infection [103]. Benztropine was also independently

identified in two drug screens for EBOV antivirals

[100, 101]. A recent report suggests that a step after virus

attachment, but prior to viral/cell membrane fusion is tar-

geted by benztropine [104].

Clomipramine, a tricyclic antidepressant, and thiothix-

ene, a thioxanthene antipsychotic, have also been shown to

inhibit MERS-CoV and SARS-CoV infection in vitro [38].

In addition, they were found to inhibit EVD VLP entry

[101]. Both are approved clinically, and clomipramine

belongs to the WHO Model List of Essential Medicines.

Several other neurotransmitter inhibitors, astemizole,

promethazine, chlorphenoxamine, and fluspirilene, were

active against MERS-CoV and SARS-CoV in cell culture,

but we were not able to find reports on activity against

other viruses [38]. Astemizole is an H1-histamine receptor

antagonist for treating allergic rhinitis that was withdrawn

from the market. Cardiac adverse events due to drug

overdose have been reported, but are extremely rare [105].

Recently, astemizole has gained renewed interest as an

anticancer and antimalarial drug [106, 107].

Chlorphenoxamine is an antihistamine and anticholin-

ergic that is currently in preclinical trials for malaria.

Fluspirilene is an approved antipsychotic for treatment of

schizophrenia. It is a known autophagy inducer [108].

Autophagy is a cellular degradative pathway that viruses

exploit for their propagation [109]. Modulators of autop-

hagy may perturb MERS-CoV or SARS-CoV infection,

and investigation of their broad-spectrum potential for the

treatment of coronaviral infections would be interesting

[110].

2.2.7 Nucleic Acid Synthesis Inhibitors

Several RNA/DNA synthesis inhibitors have broad-spec-

trum activity against viruses including SARS-CoV and

MERS-CoV [38, 80, 111–114]. Inosine monophosphate

dehydrogenase (IMPDH) inhibitors such as ribavirin,

mycophenolic acid, and mizoribine inhibit an important

step in de novo synthesis of nucleic acids although the

potency of these drugs against viruses varies. Ribavirin has

been used in combination with IFN in the clinic for treat-

ment of viral infections such as hepatitis C. Treatment

regimens with ribavirin are well characterized and have

been used in SARS and MERS patients with mixed results

(see 2.1). Ribavirin weakly inhibits MERS-CoV in vitro,

and conflicting data have been reported for the activity of

ribavirin against SARS-CoV [80, 111, 115]. Many of the

studies on ribavirin were performed in Vero cells that

reportedly have a defect in ribonucleoside uptake, which

could explain lack of activity for ribavirin in these cells

[116]. Another coronavirus, mouse hepatitis virus (MHV),

becomes sensitive to ribavirin when its exoribouclease

activity is inactivated. In presence of exoribonuclease,

ribavirin does not inhibit MHV replication [117]. The

MHV exoribonuclease has been suggested to function as a

‘proofreading’ viral enzyme that is necessary for high-fi-

delity replication of MHV. Similarly, the exoribonuclease

activity of MERS-CoV and SARS-CoV could possibly

counteract inhibitory activity of ribavirin.

Mycophenolic acid (MPA), an immunosuppressant used

to prevent organ rejection, has broad-spectrum antiviral

activities, and antifungal, antibacterial, anticancer, and

antipsoriatic properties [45, 118, 119]. Although MPA has

weak inhibitory activity against SARS-CoV in vitro, it has

promising activity against MERS-CoV [80, 112]. A

potential alternative to MPA, the prodrug mycophenolate

mofetil, has improved oral bioavailability [120].

Mycophenolate mofetil evaluated in the common mar-

moset model of MERS did not reduce disease manifesta-

tions compared to that observed in control subjects [121].

However, the MERS marmoset model does not recapitulate

human disease due to its rapid onset and pathology asso-

ciated with exposure methods [122]. Mizoribine, an

approved immunosuppressant in organ transplantation with

limited adverse side effects, has shown in vitro activity

against HCV and bovine viral diarrhea virus (BVDV), and

was considered as an alternative to ribavirin/IFN combi-

nations for treatment of HCV infections [123]. In vivo

analysis of ribavirin and other IMPDH inhibitors in SARS-

CoV-infected mice have suggested that these agents would

be of limited benefit [111].

The chemotherapeutic gemcitabine, has shown in vitro

activity against MERS-CoV and SARS-CoV [38]. The

drug’s anti-cancer mechanism is attributed to its ability to

inhibit ribonucleotide reductase essential for de novo

pyrimidine biosynthesis. Gemcitabine has been shown to

suppress influenza virus RNA transcription and replication

by targeting ribonucleotide reductase and showed anti-

retroviral activity in vivo in the mouse model for murine

leukemia virus [124, 125].

1950 J. Dyall et al.

Page 17: Middle East Respiratory Syndrome and Severe Acute ... · The outbreaks of Middle East respiratory syndrome (MERS) and severe acute respiratory syndrome (SARS) were caused by emerging

2.2.8 Protease Inhibitors

MERS-CoV and SARS-CoV require activation of their

envelope glycoproteins by host proteases for cell entry by

the endosomal or the non-endosomal pathways. Inhibitors

of host cell proteases are being investigated as possible

antivirals [126]. The serine protease TMPRRSS2 mediates

entry via the non-endosomal pathway for both MERS-CoV

and SARS-CoV [50, 127–129]. Camostat mesylate, which

has been used in the treatment of chronic pancreatitis,

inhibits TMPSSR2-mediated glycoprotein activation of

MERS-CoV, SARS-CoV, and influenza virus

[126, 130–132]. K11777, a cysteine protease inhibitor, is in

clinical development for treating parasitic infections.

K11777 has broad-spectrum activity against coronaviruses

(MERS-CoV, SARS-CoV, HCoV-229E), filoviruses

(EBOV, Marburg virus), and paramyxoviruses (Nipah

virus) [133]. Interestingly, Zhou et al. [133] demonstrated

that Camostat and K11777 had inhibitory activity against

SARS-CoV whereas EBOV was only inhibited by K11777,

suggesting differential host protease requirements for these

viruses [133]. E-64-D, an inhibitor of an endosomal cys-

teine protease currently in Phase III trials for the treatment

of muscular dystrophy, inhibits both MERS-CoV and

SARS-CoV in vitro [38]. E-64-D also inhibits filovirus cell

entry [134, 135]. The dependency of viruses for specific

serine or cysteine host proteases must be considered in the

selection of protease inhibitors for antiviral therapeutic

applications. Therefore, an increased understanding of the

relationship between host proteases and viral pathogenesis

will determine the most effective treatment options for

viral infections.

Lopinavir was identified as an inhibitor of MERS-CoV

and SARS-CoV in vitro, and time-of-addition experiments

indicate that the drug acts at an early stage of viral entry

[36]. Lopinavir, an inhibitor of the HIV protease, is used

clinically for the treatment of HIV infections [136]. It is

given in combination with ritonavir, an inhibitor of cyto-

chrome P450 3A4, to increase blood concentrations

because of the low bioavailability of lopinavir [136].

Lopinavir also inhibits human papilloma virus and is cur-

rently under development for the topical treatment of cer-

vical cancer [137]. Treatment with lopinavir/ritonavir

resulted in reduced mortality in a NHP model of MERS

[121]. Lopinavir has been shown to target the main pro-

tease (Mpro) of SARS-CoV [138]. However, lopinavir has

also been shown to act on other intracellular processes that

are involved in coronavirus replication. Additional studies

are needed to fully understand the mechanism of action of

lopinavir involving cellular proteases. During the 2003

SARS outbreak, patients in open clinical trials were treated

with lopinavir/ritonavir in combination with ribavirin had a

milder disease course and reduction in fatality rate com-

pared to that observed with historical controls [27, 28].

2.2.9 Protein Synthesis Inhibitors

Three protein synthesis inhibitors with activity against

coronaviruses were identified, emetine, anisomycin and

omacetaxine mepesuccinate [38, 139]. Emetine, a natural

plant alkaloid, and anisomycin, an antibiotic, both inhibit

protein elongation and were identified as anti-protozoals

[140, 141]. While emetine is approved for amoebiasis

treatment, anisomycin did not move beyond clinical trials

[141, 142]. Dehydroemetine, a synthetic emetine deriva-

tive, has fewer side effects and is available as an investi-

gational new drug [143, 144]. Anisomycin was originally

discovered as a peptidyl transferase inhibitor, but also

activates the MAP kinase signaling pathway [145]. In

addition to activity against MERS-CoV and SARS-CoV,

emetine and anisomycin inhibit the animal picornavirus

encephalomyocarditis virus [146]. Anisomycin has in vitro

activity against poliovirus [147]. Omacetaxine mepesuc-

cinate, a plant-derived alkaloid, is an anticancer therapeutic

that received FDA approval in 2012 for the treatment of

chronic myeloid leukemia. Omacetaxine inhibits MERS-

CoV, bovine coronavirus, human enteric coronavirus and

hepatitis B virus [139, 148]. In spite of omacetaxine broad

spectrum anti-coronavirus activity, the drug had no activity

against SARS-CoV [38]. Drugs that inhibit coronaviruses

by targeting protein synthesis may have potential in the

development of combination therapies with drugs that

target other antiviral pathways.

2.2.10 Selective Estrogen Receptor Modulators

Recent investigations have demonstrated the potential of

estrogen receptor (ER) antagonists for repurposing as anti-

coronavirus compounds [38]. For example, toremifene

citrate and tamoxifen citrate with activity against SARS-

CoV and MERS-CoV were developed and approved as

anticancer therapeutics. Both drugs have shown activity

against HCV replication in vitro [149, 150]. Mechanistic

studies revealed that the ER is functionally associated with

HCV replication [150]. ER promotes the interaction

between the HCV replication complex and the HCV

polymerase NS5B. ER–mediated regulation of HCV gen-

ome replication is abrogated by tamoxifen.

Toremifene and tamoxifen also effectively inhibit

EBOV infection in vitro [151]. However, in contrast to

HCV, mechanistic studies have shown that toremifene-

mediated EBOV inhibition is independent of the ER

pathway as toremifene was still active against EBOV in

cells that did not express ER [151, 152]. Toremifene acts at

a late step of virus entry after internalization of EBOV and

Therapeutics for MERS-CoV and SARS-CoV 1951

Page 18: Middle East Respiratory Syndrome and Severe Acute ... · The outbreaks of Middle East respiratory syndrome (MERS) and severe acute respiratory syndrome (SARS) were caused by emerging

may prevent fusion between the viral and endosomal

membranes [151–153]. Based on the chemical structure,

toremifene is a cationic amphiphilic drug (CAD) that is

known to be lysosomotropic and could affect endosomal

processes during virus entry [151, 154]. Treatment with

toremifene led to 50% survival of EBOV–infected mice

confirming that this drug has an effect in vivo as well

[151].

In terms of clinical application, toremifene and tamox-

ifen have good bioavailability, safety and tolerability pro-

files combined with a long history of use in the clinic.

However, prolongation of the QT interval has been noted

for toremifene and should not be prescribed to patients with

congenital or acquired long QT syndrome, uncorrected

hypokalemia or uncorrected hypomagnesemia [155].

Tamoxifen can increase uterine malignancies, stroke and

pulmonary embolism in women with ductal carcinoma

in situ or at high risk for breast cancer [156]. Despite these

side effects, the drugs may have substantial value for short-

term treatment of acute coronaviral infections. Advanced

patient studies and careful evaluation of the pharmacoki-

netic profiles may facilitate dosing strategies that limit the

risk of adverse events.

2.2.11 Sterol Metabolism Inhibitors

Two sterol synthesis inhibitors, terconazole and triparanol,

have shown activity against MERS-CoV and SARS-CoV

[38]. Studies with virus-like particles (VLPs) have

demonstrated that terconazole inhibits coronavirus cell

entry, including MERS-CoV and SARS-CoV. The sterol

synthesis pathway has been shown to be required for

infection by several viruses including HCV [157, 158].

Terconazole, approved for vaginal yeast infections, can be

administered orally, topically or by suppository. Triparanol

was approved for lowering plasma cholesterol, but was

withdrawn due to numerous side effects. Both are CADs

that induce accumulation of cholesterol in late endosomes

and have been shown to inhibit EBOV entry [152].

2.3 Drugs in Development

2.3.1 Potential Targets for Inhibition of MERS-CoV

and SARS-CoV

In addition to drug repurposing, development of novel

antiviral countermeasures is needed for emerging coron-

aviruses. To this end, design or development strategies

have targeted the viral replication cycle and host pathways

essential for viral replication (Table 4). Two nucleoside

inhibitors of viral RNA-dependent RNA polymerases, GS-

5734 and BCX4430, have potential as broad-spectrum

antivirals [159, 160]. Both drugs are active against MERS-

CoV and SARS-CoV in cell culture, but in vivo efficacy

remains to be investigated. In addition, a new class of

nucleosides with a flexible purine base has anti-coronaviral

activity, and further optimization could generate potent

inhibitors of the coronaviral polymerase [161]. The surface

glycoprotein (S) of SARS-CoV and MERS-CoV and other

coronaviruses consists of two domains: S1, containing the

receptor-binding domain (RBD) needed for extracellular

binding; and S2, containing the fusion peptide needed for

membrane fusion and release. Endocytosis of SARS-CoV

is facilitated by the binding of RBD with the angiotensin

converting enzyme 2 (ACE2) receptor on host cells.

Membrane-bound cathepsin L cleaves the S protein

revealing the S2 fusion protein, which fuses with the

membrane and releases the viral RNA. Inhibitors of

cathepsin L, the ACE2–SARS–S1 complex, or the S2

fusion peptide could be suitable targets to inhibit SARS-

CoV entry [162]. Results from recent studies have identi-

fied inhibitors of viral entry, viral proteases, and helicases

that potently inhibit both MERS-CoV and SARS-CoV

[162]. Proteases, such as papain-like protease and 3C-like

protease, could also be useful as antiviral targets for drug

development as they are required for cleaving non-struc-

tural proteins for viral maturation. Most protease inhibitors

are ‘‘suicide’’ protease inhibitors that bind to the target

irreversibly. However, reversible protease inhibitors may

have greater potential as they are less toxic and better

tolerated [113]. Recent studies with helicase inhibitors

show that three domain targets, N-terminal metal binding

domain, a hinge domain, and a NTP/helicase domain, have

potential for the development of new drugs [163].

2.3.2 RNA Interference

Directed RNA interference (RNAi) presents a powerful

approach for the development of novel virus-specific

therapeutics based on gene silencing [164]. Recent studies

have shown that small interfering RNAs (siRNAs) or short

hairpin RNAs can inhibit expression of viral genes and

thereby block the replication of SARS-CoV in cultured

cells [165–172]. Intranasal delivery of a combination of

small interfering RNA (siSC2-5) targeting SARS-CoV

open reading frame 1 and S protein decreased SARS

pathogenesis in NHPs [173]. Potential RNAi candidates

identified through computational modeling for MERS-CoV

require further in vitro and preclinical investigation [174].

Several RNAi therapeutics for the treatment of viral

infections have entered clinical trials including TKM-

Ebola, a siRNA/lipid nanoparticle platform targeting

EBOV [164, 175, 176]. This technology has great potential

for therapeutics for emerging viruses as viral genome

1952 J. Dyall et al.

Page 19: Middle East Respiratory Syndrome and Severe Acute ... · The outbreaks of Middle East respiratory syndrome (MERS) and severe acute respiratory syndrome (SARS) were caused by emerging

sequencing can now be completed in a very short time

frame that is crucial in situations when an epidemic of a

novel emerging viral infection unfolds. The main obstacle

for RNAi strategies lies in the identification of suitable vi-

ral targets and in the delivery efficiency of nucleic acids to

target cells in vivo.

2.3.3 Peptide Entry Inhibitors

Peptides share common physicochemical properties with

CADs that facilitate interaction with cell membranes and

interference with the fusion of cellular and viral mem-

branes during virus entry. Researchers are making progress

in defining the mechanism of action of peptide entry

inhibitors of enveloped viruses such as enfuvirtide, an

approved HIV inhibitor [177]. Enfuvirtide is a 36-residue

peptide derived from the amphipathic loop/C-helix heptad

repeat domain of HIV gp41. A rational approach based on

scanning fusion protein sequences for amphipathic

sequences has led to the discovery of additional peptide

inhibitors for other viruses including MERS-CoV and

SARS-CoV [178–180]. Chemical modifications of peptides

have increased their in vivo stability and bioavailability,

improving their potential for clinical applications as novel

broad-spectrum viral entry inhibitors [181–183].

Table 4 Drugs in development for the treatment of Middle East respiratory syndrome (MERS) or severe acute respiratory syndrome (SARS)

Viral/cellular target Drug class Drug

MERS-CoV

3C-like protease Benzotriazole esters CE-5 [284]

Papain-like protease Thiopurines 6-Thioguanine, 6-mercaptopurine [113]

Helicase Triazole SSYA10-001 [162]

RNA-dependent RNA

polymerase

Nucleotide prodrug GS-5734 [159]

RNA-dependent RNA

polymerase

Nucleoside analog BCX4430 [160]

Membrane-bound

RNA synthesis

Small molecule inhibitor K22 [285]

Furin inhibitor Small molecule inhibitor Decanoyl-RVKR [286]

SARS-CoV

3C-like protease Benzotriazole esters CE-5 [284]

3C-like protease Anilides Peptide nitroanilides [287]

3C-like protease C2-symmetric inhibitors containing diol cores TL-3 [288]

3C-like protease Pyrazole analogs Pyrazolones [289]

3C-like protease Serine inhibitor Trifluoromethyl ketones [290]

3C-like protease Serotonin receptor antagonist Cinanserin [291]

3C-like protease Zinc-conjugated inhibitor JMF 1586 [292]

Papain-like protease Thiopurines 6-Thioguanine, 6-mercaptopurine [113]

Helicase Triazole SSYA10-001 [162]

Helicase Bananin derivatives Vanillinbananin, Idobananin [163]

NTPase/Helicase Aryl diketoacids Dihydroxychromone and

hydroxychromone derivatives

ADK analogs [293]; 2-(3-iodobenzyloxy)-6-(3-chlorobenzyloxy)-

5-hydroxychromone [294]

RNA-dependent RNA

polymerase

Nucleoside analogs BCX4430 [160], 4-aza-7,9-dideazaadenosine C-nucleosides [295],

fleximer nucleoside analogs [161]

Cathepsin L cellular

protease

Small molecule inhibitor Oxocarbazate [296], SSAA09E1 [297]

ACE2–SARS–S1

complex

Small molecule inhibitor SSAA09E2 [297]

S2-cell membrane

fusion

Small molecule inhibitor SSAA09E3 [297]

ACE2–SARS–S1

complex

Small molecule inhibitor NAAE [298]

ACE2 angiotensin converting enzyme, MERS-CoV Middle East respiratory syndrome coronavirus, S1 spike protein 1 domain, S2 Spike protein 2

domain

Therapeutics for MERS-CoV and SARS-CoV 1953

Page 20: Middle East Respiratory Syndrome and Severe Acute ... · The outbreaks of Middle East respiratory syndrome (MERS) and severe acute respiratory syndrome (SARS) were caused by emerging

2.4 Antibody Therapy

The success of palivizumab for treatment of respiratory

syncytial virus infection has reinvigorated efforts to

develop monoclonal antibody-based therapeutics for

infectious diseases [184]. ZMapp, a monoclonal antibody

cocktail targeting EBOV, has been tested in NHPs with

success and was moved to Phase I and II clinical trials in

humans during the EVD epidemic [185, 186]. Similarly, a

monoclonal antibody against Hendra virus has been

administered to humans on a ‘‘compassionate use’’ basis

[187–189]. These examples demonstrate the potential for

antibody therapy to combat emerging/re-emerging viruses,

and similar strategies have been pursued by multiple

groups for development of antibodies to MERS-CoV

[190–197]. Monoclonal antibodies to MERS-CoV have

been sourced from humanized mice libraries or human

antibody libraries [192, 194, 197, 198]. Antibodies target

the S RBD, S1, or S2 subunits, and have demonstrated

efficacy in animal models as reviewed in Ying et al. [199].

Monoclonal antibody therapy can impart a selective pres-

sure for generation of resistant viruses. Although mutants

escaping monoclonal antibody pressure tend to be less fit,

analysis of the emergence of mutations that confer resis-

tance to the monoclonal antibody will need to be

performed.

An alternative to monoclonal antibody therapy is poly-

clonal antibody therapy using convalescent sera (sera

sourced from a nonhuman or humanized animal). Poly-

clonal antibodies provide an advantage over monoclonal

antibodies in that escape mutants are less likely to emerge

[200, 201]. Convalescent sera have been recommended for

MERS, and a Phase II interventional clinical trial is

ongoing to determine efficacy [198]. However, the avail-

ability of a suitable donor subject presents a significant

complication for this approach. Nonhuman animal sera has

been considered, but safety concerns limit this option.

Fractionation of nonhuman sera is an alternative; however,

antibody-mediated clearance can be limited due to failure

of the human Fc receptors to recognize the antibody heavy

chain.

An alternative is de-speciating the antibodies by using

only the Fab antibody fragment; however, cost and suffi-

cient material may make mass production of Fab fragments

difficult. The use of sera from humanized mice or other

small laboratory animals is complicated by sample acqui-

sition/volume restraints. Larger laboratory animals may

provide a potential alternative. SAB Biotherapeutics has

developed a trans-chromosomic bovine platform for the

generation of human IgG antibodies [201]. Vaccination of

trans-chromosomic cattle with S protein nanoparticles or

inactivated, whole virus generated fully humanized

polyclonal antibodies that demonstrated efficacy in the Ad-

DPP4 murine model of MERS.

3 Lessons Learned

One of the most important lessons regarding antiviral drug

development is that both highly specific and broad-spec-

trum antivirals bring unique advantages to the table.

Antiviral agents can fall anywhere in the spectrum between

‘‘broad-spectrum’’ and ‘‘highly specific’’ [202]. A drug that

targets a specific virus or virus family will have narrow

activity, high potency, and low toxicity; however, such a

drug may also promote resistance [202]. In contrast, broad-

spectrum antivirals typically target a host factor or path-

way, and often these agents have higher toxicities, lower

potencies, and delayed treatment effects. However, the

selective pressure for resistance is often lower with broad-

spectrum agents.

A large part of our knowledge on antiviral development

stems from the studies of chronic viral infections. Antiviral

development strategies for DNA viruses have been suc-

cessful in identifying a single drug that specifically targets

a viral protein. This strategy has been less successful for

RNA viruses. RNA viruses mutate at a higher rate than

DNA viruses resulting in enhanced development of drug

resistance.

3.1 AIDS

Despite extensive efforts over the past 30 years, a thera-

peutic or prophylactic HIV vaccine has remained elusive.

Antiviral agents are the only available treatments for AIDS.

Over 25 antivirals belonging to 6 different drug classes

targeting different stages of viral replication are available

(e.g. reverse transcriptase, protease, fusion, entry, inte-

grase) [203]. Combination treatments with 2 to 3 drugs are

effective and result in a sustained virologic response [204].

Two aspects have been found to be important for avoiding

resistance: (1) selecting drugs with at least two different

targets, and (2) selecting drugs that belong to different

chemical classes. These considerations may also apply for

drug combinations with synergistic effects against MERS-

and SARS-CoV.

3.2 Hepatitis C

Broad-spectrum antiviral therapies can be of great value for

treating emerging infections when it takes time to develop

direct-acting antivirals. For treatment of chronic hepatitis

C, clinicians have depended on IFN and ribavirin for a

number of years [205]. Eventually, IFN and ribavirin

combination was replaced by very effective fixed-

1954 J. Dyall et al.

Page 21: Middle East Respiratory Syndrome and Severe Acute ... · The outbreaks of Middle East respiratory syndrome (MERS) and severe acute respiratory syndrome (SARS) were caused by emerging

combination therapies using direct-acting antivirals that

target multiple steps of the HCV life cycle [206]. IFN and

ribavirin contribute significantly to the treatment of viral

infections for which no direct-acting antivirals exist.

However, they have major side effects [207]. More options

for broad-spectrum antivirals with improved safety profiles

would be beneficial for use for emerging coronavirus

infections.

3.3 Influenza

Influenza viruses are characterized by a high mutation rate

of the RNA genome. As available vaccines may not be

protective against a novel pandemic strain, antiviral agents

are considered an essential component for preparedness.

Combinations of direct-acting antivirals are under evalua-

tion for additive or synergistic effects and prevention of

resistance [208, 209]. One triple combination (oseltamivir,

amantadine and ribavirin) is synergistic and prevents

resistance in vivo [210, 211], highlighting the potential of

combinatorial therapy.

3.4 Ebola Virus Disease

The recent epidemic of EVD inWestern Africa has renewed

the urgency for development of treatments against emerging

viruses. Although vaccines and direct-acting antiviral treat-

ment are under investigation, none are approved for clinical

use [212–216]. The WHO prioritized a panel of drugs

approved for other indications that were considered for

repurposing under FDA’s Emergency Use Authorization

[62]. Two of these drugs also have activity against MERS-

CoV and SARS-CoV, amodiaquine (antimalarial agent) and

toremifene citrate (breast cancer treatment).

Additional broad-spectrum antiviral agents (including

repurposed drugs) should be a top priority for future

emerging infections including coronavirus infections. A

panel of broad-spectrum drugs that have been carefully

validated for efficacy and safety and that could be used in

combination would supply a minimum of protection for

patients and healthcare workers at outbreak locations. This

panel of drugs could be used in situations of a known re-

emerging pathogen for which specific antiviral agents and

vaccines has not been approved or of an unknown novel

pathogen that could arise.

4 Gaps in Knowledge and Future Outlook

4.1 Animal Models of MERS and SARS

Effective development of countermeasures depends on

developing appropriate animal models that uniformly

recapitulate human disease progression and severity of

pathological manifestations. As with most animal models

of human disease, no one animal model fully reflects SARS

or MERS, therefore researchers are faced with exploring

several small animal models or choosing the ‘‘best-fit’’

model. To date, animal models do not fully recapitulate

human disease, thus animal models of MERS and SARS

need further refinement. Many small animal models have

been evaluated as potential MERS and SARS models

including mice, hamsters, and ferrets for MERS and Syrian

hamsters, and guinea pigs for SARS [217–219]. Four

murine models have been reported for MERS. The first

model that demonstrated promise involved transduction of

the respiratory tract with the putative MERS-CoV receptor,

human DPP4 (or CD26) [220]. The major indicator of

disease in this model is viral load in the lung at 4 days post-

inoculation. Although clinical signs of disease, including

weight loss, were limited in this model, it has been used for

pathogenesis countermeasure studies [221, 222]. Lethal,

disseminated MERS infection has been demonstrated in

transgenic mice expressing human DPP4 [223]. Inflam-

matory processes were observed in the brains of these mice

in contrast to human disease in which CNS involvement

has not been reported. A transgenic mouse MERS model

was developed in which the mouse DPP4 gene was

replaced by the human DPP4 gene under control of the

endogenous mouse DPP4 promoter. Using this model,

MERS-CoV-infected mice developed lung pathology

[194]. In addition, administration of human monoclonal

antibodies against the spike protein in these transgenic

mice provided protection against MERS-CoV infection

[194]. Clustered regularly interspaced short palindromic

repeat-CRISPR-associated protein 9 (CRISPR-CAS9) gene

editing technology was used to modify the mouse DPP4 to

match human DPP4 by altering amino acids at positions

288 and 330. Interestingly, wild type virus infection of

these mice did not result in an improved model of MERS.

However, serial passage of MERS-CoV resulted in MERS-

CoV-15. Intranasal exposure of MERS-CoV-15 in

288/330, mice led to weight loss and a severe respiratory

disease that included ARDS-like signs and reduced pul-

monary function [224].

MERS-CoV infection of rabbits has also been evaluated

as a model for MERS. Haagmans et al. [225] demonstrated

that MERS-CoV infected rabbits did not develop obvious

clinical signs, but infectious virus could be detected in the

upper respiratory tract [225]. Furthermore, epithelial cells

of the bronchioles and terminal bronchioles respiratory

tract were positive for MERS-CoV by immunohistochem-

istry and in-situ hybridization, which reflects tissue tropism

in human disease [225]. Using the rabbit model, Houser

et al. [191] demonstrated that human monoclonal antibody

336 given pre-exposure reduced viral RNA lung titer at

Therapeutics for MERS-CoV and SARS-CoV 1955

Page 22: Middle East Respiratory Syndrome and Severe Acute ... · The outbreaks of Middle East respiratory syndrome (MERS) and severe acute respiratory syndrome (SARS) were caused by emerging

3 days post-exposure, but not when given post-exposure

[191].

Due to phylogenetic similarities with humans, NHP

models of disease have long been considered as necessary

for evaluation of countermeasures to infectious diseases.

Rhesus monkey and common marmoset have been evalu-

ated as potential models for MERS. Following intratracheal

instillation of MERS-CoV in rhesus monkey models, lung

pathology was observed [122, 226–228]. Experiments

using rhesus monkeys have indicated that they develop

limited systemic disease and a transient respiratory disease.

Radiologic evaluations have indicated inflammatory infil-

trates that develop shortly after exposure. Analysis of lung

tissues by reverse transcriptase- quantitative polymerase

chain reaction indicated virus replication in the lung.

Similar to MERS, African green monkeys (AGMs),

rhesus monkeys, cynomolgus monkeys, and common

marmosets have been identified as potential models for

SARS [229]. Smits et al. [230] compared SARS CoV

infection in young AGMs to cynomolgus monkeys, they

observed that neither species developed clinical signs

during a 4-day experiment [230]. Gross pathology indi-

cated multi-focal pulmonary consolidation with consoli-

dated grey-red firm lungs. These lesions affected 30% of

the lungs in one subject. By comparison, the cynomolgus

monkeys developed small patchy macroscopic lesions.

Similar to MERS-CoV infection of NHPs, viral load

decreases from exposure day to day 4. A comparison

between AGMs, rhesus monkeys, and cynomolgus mon-

keys further support AGMs as the best available NHP

model for SARS [231]. AGMs developed the highest viral

load and most disease when compared to cynomolgus and

rhesus monkeys. Lethal disease was not observed in any of

these species; therefore, further development of the SARS

model is warranted since lethal respiratory tract disease

was the hallmark of SARS.

As an alternative to Old World NHPs, many groups have

employed marmosets as models of human infectious dis-

ease [232–234]. Common marmosets have been evaluated

as a MERS model [121, 122, 235]. These studies have

demonstrated that common marmosets develop disease

following exposure to MERS-CoV as shown by

histopathological analysis, radiological analysis, and RT-

qPCR. However, variable results have been reported, and

exposure methodology can impact disease progression.

Therefore, mock-infected groups must be included to

account for pathological artifacts. The virus-specific

pathology could be quantified using computed tomography,

and future experiments using large group sizes could be

used for countermeasure evaluation. Greenough et al. [236]

performed a serial euthanasia study of SARS-CoV infected

marmosets [236]. Subjects were intratracheally exposed

with SARS-CoV. They observed mild inconsistent clinical

signs of disease. Viral loads peaked at day 4 post-infection.

Histopathology indicated interstitial pneumonitis with

multinucleated syncytia that were described as mild and

not observed in all late time-point subjects. Overall, further

research is needed to develop animal models of SARS that

reflect human disease presentation.

4.2 Combinations with Synergy

Drugs with repurposing potential discussed here (Table 3)

have the advantage of easy access, availability and

decreased cost of development and provide a wide array of

options for combination studies. The pharmacological

knowledge available for such compounds may also reduce

concerns regarding adverse effects in patients. The gener-

ation of a translational database encompassing pharmaco-

dynamics data and infectious disease biology data has been

proposed and would greatly facilitate decision making to

pursue new drug combinations [237]. Many of the drugs

have potential for broad-spectrum antiviral activity and

have already been in clinical use for treating other viral

infections. As novel drugs in development move from the

pre-clinical to clinical phase, they also become available

for combination therapy. Care should be taken with the

pharmacological evaluation of each combination to avoid

possible contraindications of the drugs with regards to

disease or adverse effects. Novel broad-spectrum replica-

tion inhibitors, such as GS-5734 (Gilead Sciences, in Phase

I clinical trial), immunomodulators (nitazoxanide; steroids;

statins) along with direct-acting antiviral agents for coro-

naviruses that are in development represent interesting

partners for combinations. Combinations can involve

broad-spectrum versus specific antiviral agents; drugs with

different mechanism of action; or drugs that target different

steps of the viral life cycle. Identifying one or more potent

combinations with activity in an animal model would

greatly increase preparedness for the next coronavirus

outbreak.

4.3 Structure-Based Drug Design

Elucidation of the crystal structure of viral proteins has led

to novel approaches for rational drug design. Rational

design investigations using protein structure information

and in silico screening for affinity to active sites of viral

proteins holds promise. HIV-1 protease inhibitors have

been one of the big successes of rational drug design. Only

6 years after the publication of the HIV-1 protease struc-

ture, saquinavir was developed in record time from bench

to bedside and was licensed for use against AIDS in 1995

[238–240]. In total, six antivirals against the HIV-1 pro-

tease were designed and approved between 1995 and 2000.

Similarly, for HCV, computer–aided approaches based on

1956 J. Dyall et al.

Page 23: Middle East Respiratory Syndrome and Severe Acute ... · The outbreaks of Middle East respiratory syndrome (MERS) and severe acute respiratory syndrome (SARS) were caused by emerging

the known crystal structure of a viral protein have suc-

cessfully guided the design and synthesis of inhibitors for

the HCV NS3/NS4A proteases such as the peptidomimet-

ics, telaprevir and boceprivir [241–243].

Due to the power of computational modeling using

crystal structures from known coronaviruses, the crystal

structures for the viral proteases, Mpro and PLpro, of SARS-

CoV and MERS-CoV were determined relatively quickly

[244]. These structures have already been used for the

discovery of inhibitors with high binding affinity to the

active site of the proteases. The structures of additional

MERS-CoV or SARS-CoV viral proteins have yet to be

determined and would offer additional viral targets for drug

discovery.

Structural design can help with development of inhi-

bitors in preparing for future outbreaks of yet unknown

emerging coronaviruses. Based on the potential of zoo-

notic transmission of coronaviruses from bats to humans,

crystal structures for the main protease (Mpro) of dif-

ferent bat coronavirus families have been proposed for

screening and identifying broad-spectrum antiviral

agents [244]. Proof-of-principle was shown for the novel

protease inhibitor SG85, which inhibits the bat coron-

avirus HKU4 [244]. In 2012, SG85 was quickly identi-

fied as an inhibitor of MERS-CoV through in silico

docking studies with the Mpro of MERS-CoV and the bat

coronavirus HKU4.

4.4 Cationic Amphiphilic Drugs: A Novel Class

of Antiviral Agents?

Several drugs (e.g. toremifene citrate, terconazole,

Fig. 3) belong to a group of compounds termed cationic

amphiphilic drugs (CADs) [100, 151, 152, 154, 245].

Phenothiazines (e.g. fluphenazine chlorpromazine,

Fig. 3) are CADs that have been shown to inhibit HCV

entry at virus-host cell fusion by intercalating into the

cholesterol-rich domains of the host cell membrane and

increasing membrane fluidity [97]. Drugs that act

through this mechanism may present an interesting new

class of broad-spectrum antivirals. CADs are known to

be lysomotropic and accumulate in acidic compartments

where their tertiary amine groups are protonated. The

compounds act as mild bases and can neutralize the low

pH of the acidic environment of endo/lysosomes. CADs

can intercalate into membranes, alter the biophysical

properties of membranes and thereby could potentially

interfere with fusion of virus with the endo/lysosomal

membrane. The concept of interfering with virus entry

and budding through physicochemical properties of

drugs is intriguing. Many viruses would be susceptible to

this type of inhibition and CADs could be used as broad-

spectrum antiviral agents. Detailed structure-activity

relationship studies on CADs will be required to deter-

mine the chemical core structures and physicochemical

properties important for this type of antiviral agent.

Future investigations regarding the conservation of this

mechanism of action to coronaviruses, as well as other

emerging viruses, are warranted.

4.5 Analogs of Developed Drugs

Approved drugs with activity against MERS-CoV or

SARS-CoV could be used as lead compounds for further

antiviral drug development. Pharmaceuticals usually have

undergone multiple rounds of structure-activity relation-

ship studies generating analogs to improve drug activity

against the original indication or target. The analogs of

drugs with activity against MERS-CoV and SARS-CoV

may still be available and could be screened to identify

other analogs with increased antiviral activity. Although

drug analogs would have to go through the full licensure

process, there would be little to no added initial cost

associated with producing the structure-activity relation-

ship compounds. Recycling old analogs is one approach

that may have value for developing novel drugs for viral

infections.

5 Conclusion

A more streamlined process is needed for development of

effective treatment measures for emerging and re-emerging

pathogens. The availability of a panel of approved broad-

spectrum drugs would clearly be beneficial as they could be

used for treating disease symptoms and reducing morbidity

until more specific acting antivirals and vaccines are

developed.

A large number of potential drugs and therapeutics for

the treatment of MERS and SARS have been discussed.

The greatest challenge will be how best to down-select and

evaluate the different approaches. As we have learned from

drug development for AIDS and hepatitis, alleviating dis-

ease symptoms and increasing life span may be a more

achievable goal rather than looking for a treatment that will

provide complete recovery. Broad-spectrum antivirals,

specific antivirals, and immune modulators each have an

important role in treating viral infections either individu-

ally or in combination. Effective communication between

the different institute partners (government, industry, aca-

demic; national, and international partners) is essential.

Combining these drug discovery efforts will increase the

chance of having one or more potential therapeutic agents

at an advanced development stage by the time another

outbreak of an emerging coronavirus occurs.

Therapeutics for MERS-CoV and SARS-CoV 1957

Page 24: Middle East Respiratory Syndrome and Severe Acute ... · The outbreaks of Middle East respiratory syndrome (MERS) and severe acute respiratory syndrome (SARS) were caused by emerging

Acknowledgements The authors thank Laura Bollinger and Jiro

Wada for providing technical writing services and graphical support,

respectively.

Compliance with Ethical Standards

The content of this publication does not necessarily reflect the views

or policies of the US Department of Health and Human Services

(DHHS) or of the institutions and companies affiliated with the

authors.

Funding This work was supported in part by the Division of Intra-

mural Research and Division of Clinical Research, National Institute

of Allergy and Infectious Diseases. This work was funded in part

through Battelle Memorial Institute’s prime contract with the US

National Institute of Allergy and Infectious Diseases (NIAID) under

Contract No. HHSN272200700016I. J.D. performed this work as

employee of Tunnell Government Services, Inc., subcontractor to

Battelle Memorial Institute (BMI). R.G. performed this work as

employee of BMI. G.G.O. performed this work as employee of MRI

Global, subcontractor to BMI.

Conflict of interest The authors have declared that they have no

conflict of interest.

References

1. Li W, Shi Z, Yu M, Ren W, Smith C, Epstein JH, et al. Bats are

natural reservoirs of SARS-like coronaviruses. Science.

2005;310(5748):676–9.

2. Lau SK, Woo PC, Li KS, Huang Y, Tsoi HW, Wong BH, et al.

Severe acute respiratory syndrome coronavirus-like virus in

Chinese horseshoe bats. Proc Natl Acad Sci USA.

2005;102(39):14040–5.

3. Revised US. surveillance case definition for severe acute res-

piratory syndrome (SARS) and update on SARS cases—United

States and worldwide, December 2003. MMWR Morb Mortal

Wkly Rep. 2003;52(49):1202–6.

4. World Health Organization. Middle East respiratory syndrome

coronavirus (MERS-CoV)—Saudi Arabia. Disease outbreak

news. 2017. http://www.who.int/csr/don/17-january-2017-mers-

saudi-arabia/en/. Accessed 24 Jan 2017.

5. World Health Organization. Middle East respiratory syndrome

coronavirus (MERS-CoV). Fact sheet. 2017.

6. Fehr AR, Perlman S. Coronaviruses: an overview of their

replication and pathogenesis. Methods Mol Biol.

2015;1282:1–23.

7. Pasternak AO, Spaan WJ, Snijder EJ. Nidovirus transcription:

how to make sense…? J Gen Virol. 2006;87(Pt 6):1403–21.

8. Perlman S, Netland J. Coronaviruses post-SARS: update on

replication and pathogenesis. Nat Rev Microbiol.

2009;7(6):439–50.

9. Eckerle I, Corman VM, Muller MA, Lenk M, Ulrich RG,

Drosten C. Replicative capacity of MERS coronavirus in live-

stock cell lines. Emerg Infect Dis. 2014;20(2):276–9.

10. Memish ZA, Mishra N, Olival KJ, Fagbo SF, Kapoor V, Epstein

JH, et al. Middle East respiratory syndrome coronavirus in bats,

Saudi Arabia. Emerg Infect Dis. 2013;19(11):1819–23.

11. Adney DR, van Doremalen N, Brown VR, Bushmaker T, Scott

D, de Wit E, et al. Replication and shedding of MERS-CoV in

upper respiratory tract of inoculated dromedary camels. Emerg

Infect Dis. 2014;20(12):1999–2005.

12. Wang LF, Shi Z, Zhang S, Field H, Daszak P, Eaton BT. Review

of bats and SARS. Emerg Infect Dis. 2006;12(12):1834–40.

13. Chowell G, Abdirizak F, Lee S, Lee J, Jung E, Nishiura H, et al.

Transmission characteristics of MERS and SARS in the healthcare

setting: a comparative study. BMC Med. 2015;13:210.

14. Hunter JC, Nguyen D, Aden B, Al Bandar Z, Al Dhaheri W,

Abu Elkheir K, et al. Transmission of Middle East respiratory

syndrome coronavirus infections in healthcare settings, Abu

Dhabi. Emerg Infect Dis. 2016;22(4):647–56.

15. Zumla A, Hui DS, Perlman S. Middle East respiratory syn-

drome. Lancet. 2015;386(9997):995–1007.

16. Rasmussen SA, Watson AK, Swerdlow DL. Middle East res-

piratory syndrome (MERS). Microbiol Spectr. 2016;4(3):1–23.

17. Chan PK, Tang JW, Hui DS. SARS: clinical presentation,

transmission, pathogenesis and treatment options. Clin Sci

(Lond). 2006;110(2):193–204.

18. Zaki AM, van Boheemen S, Bestebroer TM, Osterhaus AD,

Fouchier RA. Isolation of a novel coronavirus from a man with

pneumonia in Saudi Arabia. N Engl J Med.

2012;367(19):1814–20.

19. Guery B, Poissy J, el Mansouf L, Sejourne C, Ettahar N,

Lemaire X, et al. Clinical features and viral diagnosis of two

cases of infection with Middle East Respiratory Syndrome

coronavirus: a report of nosocomial transmission. Lancet.

2013;381(9885):2265–72.

20. WHO Mers-Cov Research Group. State of knowledge and data

gaps of Middle East respiratory syndrome coronavirus (MERS-

CoV) in humans. PLoS Curr. 2013;5.

21. Arabi YM, Arifi AA, Balkhy HH, Najm H, Aldawood AS,

Ghabashi A, et al. Clinical course and outcomes of critically ill

patients with Middle East respiratory syndrome coronavirus

infection. Ann Intern Med. 2014;160(6):389–97.

22. Assiri A, Al-Tawfiq JA, Al-Rabeeah AA, Al-Rabiah FA, Al-

Hajjar S, Al-Barrak A, et al. Epidemiological, demographic, and

clinical characteristics of 47 cases of Middle East respiratory

syndrome coronavirus disease from Saudi Arabia: a descriptive

study. Lancet Infect Dis. 2013;13(9):752–61.

23. Ajlan AM, Ahyad RA, Jamjoom LG, Alharthy A, Madani TA.

Middle East respiratory syndrome coronavirus (MERS-CoV)

infection: chest CT findings. AJR Am J Roentgenol.

2014;203(4):782–7.

24. Memish ZA, Al-Tawfiq JA, Makhdoom HQ, Assiri A, Alha-

keem RF, Albarrak A, et al. Respiratory tract samples, viral

load, and genome fraction yield in patients with Middle East

respiratory syndrome. J Infect Dis. 2014;210(10):1590–4.

25. Chiou HE, Liu CL, Buttrey MJ, Kuo HP, Liu HW, Kuo HT,

et al. Adverse effects of ribavirin and outcome in severe acute

respiratory syndrome: experience in two medical centers. Chest.

2005;128(1):263–72.

26. Leong HN, Ang B, Earnest A, Teoh C, Xu W, Leo YS. Inves-

tigational use of ribavirin in the treatment of severe acute res-

piratory syndrome, Singapore, 2003. Trop Med Int Health.

2004;9(8):923–7.

27. Chu CM, Cheng VC, Hung IF, Wong MM, Chan KH, Chan KS,

et al. Role of lopinavir/ritonavir in the treatment of SARS: initial

virological and clinical findings. Thorax. 2004;59(3):252–6.

28. Chan KS, Lai ST, Chu CM, Tsui E, Tam CY, Wong MM, et al.

Treatment of severe acute respiratory syndrome with lopinavir/

ritonavir: a multicentre retrospective matched cohort study.

Hong Kong Med J. 2003;9(6):399–406.

29. Loutfy MR, Blatt LM, Siminovitch KA, Ward S, Wolff B, Lho

H, et al. Interferon alfacon-1 plus corticosteroids in severe acute

respiratory syndrome: a preliminary study. JAMA.

2003;290(24):3222–8.

30. Omrani AS, Saad MM, Baig K, Bahloul A, Abdul-Matin M,

Alaidaroos AY, et al. Ribavirin and interferon alfa-2a for severe

Middle East respiratory syndrome coronavirus infection: a ret-

rospective cohort study. Lancet Infect Dis. 2014;14(11):1090–5.

1958 J. Dyall et al.

Page 25: Middle East Respiratory Syndrome and Severe Acute ... · The outbreaks of Middle East respiratory syndrome (MERS) and severe acute respiratory syndrome (SARS) were caused by emerging

31. Spanakis N, Tsiodras S, Haagmans BL, Raj VS, Pontikis K,

Koutsoukou A, et al. Virological and serological analysis of a

recent Middle East respiratory syndrome coronavirus infection

case on a triple combination antiviral regimen. Int J Antimicrob

Agents. 2014;44(6):528–32.

32. King Abdullah International Medical Research Center. MERS-

CoV infection treated with a combination of lopinavir /ritonavir

and interferon beta-1b (MIRACLE). Bethesda: National Insti-

tutes of Health; 2017. https://clinicaltrials.gov/ct2/show/

NCT02845843. Accessed 12 July 2017.

33. Ashburn TT, Thor KB. Drug repositioning: identifying and

developing new uses for existing drugs. Nat Rev Drug Discov.

2004;3(8):673–83.

34. Regnard C, Twycross R, Mihalyo M, Wilcock A. Loperamide.

J Pain Symptom Manag. 2011;42(2):319–23.

35. Awouters F, Niemegeers CJ, Janssen PA. Pharmacology of

antidiarrheal drugs. Annu Rev Pharmacol Toxicol.

1983;23:279–301.

36. de Wilde AH, Jochmans D, Posthuma CC, Zevenhoven-Dobbe

JC, van Nieuwkoop S, Bestebroer TM, et al. Screening of an

FDA-approved compound library identifies four small-molecule

inhibitors of Middle East respiratory syndrome coronavirus

replication in cell culture. Antimicrob Agents Chemother.

2014;58(8):4875–84.

37. Chertow DS, Uyeki TM, DuPont HL. Loperamide therapy for

voluminous diarrhea in Ebola virus disease. J Infect Dis.

2015;211(7):1036–7.

38. Dyall J, Coleman CM, Hart BJ, Venkataraman T, Holbrook MR,

Kindrachuk J, et al. Repurposing of clinically developed drugs

for treatment of Middle East respiratory syndrome coronavirus

infection. Antimicrob Agents Chemother. 2014;58(8):4885–93.

39. Keyaerts E, Vijgen L, Maes P, Neyts J, Van Ranst M. In vitro

inhibition of severe acute respiratory syndrome coronavirus by

chloroquine. Biochem Biophys Res Commun.

2004;323(1):264–8.

40. Thome R, Lopes SC, Costa FT, Verinaud L. Chloroquine:

modes of action of an undervalued drug. Immunol Lett.

2013;153(1–2):50–7.

41. Brouwers J, Vermeire K, Schols D, Augustijns P. Development

and in vitro evaluation of chloroquine gels as microbicides

against HIV-1 infection. Virology. 2008;378(2):306–10.

42. Farias KJ, Machado PR, da Fonseca BA. Chloroquine inhibits

dengue virus type 2 replication in Vero cells but not in C6/36

cells. Sci World J. 2013;2013:282734.

43. Madrid PB, Chopra S, Manger ID, Gilfillan L, Keepers TR,

Shurtleff AC, et al. A systematic screen of FDA-approved drugs

for inhibitors of biological threat agents. PLoS One.

2013;8(4):e60579.

44. Ooi EE, Chew JS, Loh JP, Chua RC. In vitro inhibition of

human influenza A virus replication by chloroquine. Virol J.

2006;3:39.

45. Pohjala L, Utt A, Varjak M, Lulla A, Merits A, Ahola T, et al.

Inhibitors of alphavirus entry and replication identified with a

stable Chikungunya replicon cell line and virus-based assays.

PLoS One. 2011;6(12):e28923.

46. Porotto M, Orefice G, Yokoyama CC, Mungall BA, Realubit R,

Sganga ML, et al. Simulating henipavirus multicycle replication

in a screening assay leads to identification of a promising can-

didate for therapy. J Virol. 2009;83(10):5148–55.

47. Randolph VB, Winkler G, Stollar V. Acidotropic amines inhibit

proteolytic processing of flavivirus prM protein. Virology.

1990;174(2):450–8.

48. Savarino A, Boelaert JR, Cassone A, Majori G, Cauda R. Effects

of chloroquine on viral infections: an old drug against today’s

diseases? Lancet Infect Dis. 2003;3(11):722–7.

49. Raj VS, Mou H, Smits SL, Dekkers DH, Muller MA, Dijkman

R, et al. Dipeptidyl peptidase 4 is a functional receptor for the

emerging human coronavirus-EMC. Nature.

2013;495(7440):251–4.

50. Gierer S, Bertram S, Kaup F, Wrensch F, Heurich A, Kramer-

Kuhl A, et al. The spike protein of the emerging betacoronavirus

EMC uses a novel coronavirus receptor for entry, can be acti-

vated by TMPRSS2, and is targeted by neutralizing antibodies.

J Virol. 2013;87(10):5502–11.

51. Di Trani L, Savarino A, Campitelli L, Norelli S, Puzelli S,

D’Ostilio D, et al. Different pH requirements are associated with

divergent inhibitory effects of chloroquine on human and avian

influenza A viruses. Virol J. 2007;4:39.

52. Marzi A, Moller P, Hanna SL, Harrer T, Eisemann J, Stein-

kasserer A, et al. Analysis of the interaction of Ebola virus

glycoprotein with DC-SIGN (dendritic cell-specific intercellular

adhesion molecule 3-grabbing nonintegrin) and its homologue

DC-SIGNR. J Infect Dis. 2007;15(196 Suppl 2):S237–46.

53. Savarino A, Lucia MB, Rastrelli E, Rutella S, Golotta C, Morra

E, et al. Anti-HIV effects of chloroquine: inhibition of viral

particle glycosylation and synergism with protease inhibitors.

J Acquir Immune Defic Syndr. 2004;35(3):223–32.

54. Vincent MJ, Bergeron E, Benjannet S, Erickson BR, Rollin PE,

Ksiazek TG, et al. Chloroquine is a potent inhibitor of SARS

coronavirus infection and spread. Virol J. 2005;2:69.

55. Browning DJ. Toxicology of hydroxychloroquine and chloro-

quine and the pathology of the retinopathy they cause. In:

Browning DJ, editor. Hydroxychloroquine and chloroquine

retinopathy. New York: Springer Science?Business Media;

2014. p. 65–83.

56. Sperber K, Chiang G, Chen H, Ross W, Chusid E, Gonchar M,

et al. Comparison of hydroxychloroquine with zidovudine in

asymptomatic patients infected with human immunodeficiency

virus type 1. Clin Ther. 1997;19(5):913–23.

57. Sperber K, Louie M, Kraus T, Proner J, Sapira E, Lin S, et al.

Hydroxychloroquine treatment of patients with human immun-

odeficiency virus type 1. Clin Ther. 1995;17(4):622–36.

58. De Lamballerie X, Boisson V, Reynier JC, Enault S, Charrel

RN, Flahault A, et al. On chikungunya acute infection and

chloroquine treatment. Vector Borne Zoonotic Dis.

2008;8(6):837–9.

59. Tricou V, Minh NN, Van TP, Lee SJ, Farrar J, Wills B, et al. A

randomized controlled trial of chloroquine for the treatment of

dengue in Vietnamese adults. PLoS Negl Trop Dis.

2010;4(8):e785.

60. Falzarano D, Safronetz D, Prescott J, Marzi A, Feldmann F,

Feldmann H. Lack of protection against ebola virus from

chloroquine in mice and hamsters. Emerg Infect Dis.

2015;21(6):1065–7.

61. Gignoux E, Azman AS, de Smet M, Azuma P, Massaquoi M,

Job D, et al. Effect of artesunate-amodiaquine on mortality

related to Ebola virus disease. N Engl J Med.

2016;374(1):23–32.

62. World Health Organization. Categorization and prioritization of

drugs for consideration for testing or use in patients infected

with Ebola. 2015. http://www.who.int/medicines/ebola-

treatment/2015_0703TablesofEbolaDrugs.pdf?ua=1. Accessed

13 Oct 2016.

63. Brickelmaier M, Lugovskoy A, Kartikeyan R, Reviriego-Men-

doza MM, Allaire N, Simon K, et al. Identification and char-

acterization of mefloquine efficacy against JC virus in vitro.

Antimicrob Agents Chemother. 2009;53(5):1840–9.

64. Clifford DB, Nath A, Cinque P, Brew BJ, Zivadinov R, Gorelik

L, et al. A study of mefloquine treatment for progressive mul-

tifocal leukoencephalopathy: results and exploration of predic-

tors of PML outcomes. J Neurovirol. 2013;19(4):351–8.

Therapeutics for MERS-CoV and SARS-CoV 1959

Page 26: Middle East Respiratory Syndrome and Severe Acute ... · The outbreaks of Middle East respiratory syndrome (MERS) and severe acute respiratory syndrome (SARS) were caused by emerging

65. Gofton TE, Al-Khotani A, O’Farrell B, Ang LC, McLachlan RS.

Mefloquine in the treatment of progressive multifocal leukoen-

cephalopathy. J Neurol Neurosurg Psychiatry.

2011;82(4):452–5.

66. Stamnes MA, Rutherford SL, Zuker CS. Cyclophilins: a new

family of proteins involved in intracellular folding. Trends Cell

Biol. 1992;2(9):272–6.

67. de Wilde AH, Raj VS, Oudshoorn D, Bestebroer TM, van

Nieuwkoop S, Limpens RW, et al. MERS-coronavirus replica-

tion induces severe in vitro cytopathology and is strongly

inhibited by cyclosporin A or interferon-alpha treatment. J Gen

Virol. 2013;94(Pt 8):1749–60.

68. de Wilde AH, Zevenhoven-Dobbe JC, van der Meer Y, Thiel V,

Narayanan K, Makino S, et al. Cyclosporin A inhibits the

replication of diverse coronaviruses. J Gen Virol. 2011;92(Pt

11):2542–8.

69. Nagy PD, Wang RY, Pogany J, Hafren A, Makinen K. Emerging

picture of host chaperone and cyclophilin roles in RNA virus

replication. Virology. 2011;411(2):374–82.

70. Flisiak R, Horban A, Gallay P, Bobardt M, Selvarajah S,

Wiercinska-Drapalo A, et al. The cyclophilin inhibitor Debio-

025 shows potent anti-hepatitis C effect in patients coinfected

with hepatitis C and human immunodeficiency virus. Hepatol-

ogy. 2008;47(3):817–26.

71. Hopkins S, DiMassimo B, Rusnak P, Heuman D, Lalezari J,

Sluder A, et al. The cyclophilin inhibitor SCY-635 suppresses

viral replication and induces endogenous interferons in patients

with chronic HCV genotype 1 infection. J Hepatol.

2012;57(1):47–54.

72. Lawitz E, Godofsky E, Rouzier R, Marbury T, Nguyen T, Ke J,

et al. Safety, pharmacokinetics, and antiviral activity of the

cyclophilin inhibitor NIM811 alone or in combination with

pegylated interferon in HCV-infected patients receiving 14 days

of therapy. Antivir Res. 2011;89(3):238–45.

73. Barnard DL, Day CW, Bailey K, Heiner M, Montgomery R,

Lauridsen L, et al. Evaluation of immunomodulators, interferons

and known in vitro SARS-coV inhibitors for inhibition of

SARS-coV replication in BALB/c mice. Antivir Chem Che-

mother. 2006;17(5):275–84.

74. Haagmans BL, Kuiken T, Martina BE, Fouchier RA, Rim-

melzwaan GF, van Amerongen G, et al. Pegylated interferon-

alpha protects type 1 pneumocytes against SARS coronavirus

infection in macaques. Nat Med. 2004;10(3):290–3.

75. Cinatl J, Morgenstern B, Bauer G, Chandra P, Rabenau H, Doerr

HW. Treatment of SARS with human interferons. Lancet.

2003;362(9380):293–4.

76. Hensley LE, Fritz LE, Jahrling PB, Karp CL, Huggins JW,

Geisbert TW. Interferon-beta 1a and SARS coronavirus repli-

cation. Emerg Infect Dis. 2004;10(2):317–9.

77. Sainz B Jr, Mossel EC, Peters CJ, Garry RF. Interferon-beta and

interferon-gamma synergistically inhibit the replication of sev-

ere acute respiratory syndrome-associated coronavirus (SARS-

CoV). Virology. 2004;329(1):11–7.

78. Falzarano D, de Wit E, Martellaro C, Callison J, Munster VJ,

Feldmann H. Inhibition of novel beta coronavirus replication by

a combination of interferon-alpha2b and ribavirin. Sci Rep.

2013;3:1686.

79. Falzarano D, de Wit E, Rasmussen AL, Feldmann F, Okumura

A, Scott DP, et al. Treatment with interferon-alpha2b and rib-

avirin improves outcome in MERS-CoV-infected rhesus maca-

ques. Nat Med. 2013;19(10):1313–7.

80. Hart BJ, Dyall J, Postnikova E, Zhou H, Kindrachuk J,

Johnson RF, et al. Interferon-beta and mycophenolic acid are

potent inhibitors of Middle East respiratory syndrome coro-

navirus in cell-based assays. J Gen Virol. 2014;95(Pt

3):571–7.

81. Wu P, Nielsen TE, Clausen MH. FDA-approved small-molecule

kinase inhibitors. Trends Pharmacol Sci. 2015;36(7):422–39.

82. Hunter T. Protein kinases and phosphatases: the yin and yang of

protein phosphorylation and signaling. Cell. 1995;80(2):225–36.

83. Cohen P. Protein kinases—the major drug targets of the twenty-

first century? Nat Rev Drug Discov. 2002;1(4):309–15.

84. Hopkins AL, Groom CR. The druggable genome. Nat Rev Drug

Discov. 2002;1(9):727–30.

85. Josset L, Menachery VD, Gralinski LE, Agnihothram S, Sova P,

Carter VS, et al. Cell host response to infection with novel

human coronavirus EMC predicts potential antivirals and

important differences with SARS coronavirus. MBio.

2013;4(3):e00165-13.

86. Ludwig S. Disruption of virus-host cell interactions and cell

signaling pathways as an anti-viral approach against influenza

virus infections. Biol Chem. 2011;392(10):837–47.

87. Tisoncik JR, Korth MJ, Simmons CP, Farrar J, Martin TR, Katze

MG. Into the eye of the cytokine storm. Microbiol Mol Biol

Rev. 2012;76(1):16–32.

88. Coleman CM, Sisk JM, Mingo RM, Nelson EA, White JM,

Frieman MB. Abelson kinase inhibitors are potent inhibitors of

severe acute respiratory syndrome coronavirus and Middle East

respiratory syndrome coronavirus fusion. J Virol.

2016;90(19):8924–33.

89. Kindrachuk J, Ork B, Hart BJ, Mazur S, Holbrook MR, Frieman

MB, et al. Antiviral potential of ERK/MAPK and PI3 K/AKT/

mTOR signaling modulation for Middle East respiratory syn-

drome coronavirus infection as identified by temporal kinome

analysis. Antimicrob Agents Chemother. 2015;59(2):1088–99.

90. Paladin Therapeutics. Impavido (Miltefosine) capsules, for oral

use prescribing information. Wilmington: Paladin Therapeutics;

2014. https://dailymed.nlm.nih.gov/dailymed/fda/fdaDrugXsl.

cfm?setid=d6658aeb-7bc1-4eef-ad0d-0a873ddbecf5&type=

display. Accessed 13 July 2017.

91. Centers for Disease Control and Prevention. Investigational drug

available directly from CDC for the treatment of infections with

free-living amebae. MMWR Morb Mortal Wkly Rep.

2013;62(33):666.

92. Wang CH, Chung FT, Lin SM, Huang SY, Chou CL, Lee KY,

et al. Adjuvant treatment with a mammalian target of rapamycin

inhibitor, sirolimus, and steroids improves outcomes in patients

with severe H1N1 pneumonia and acute respiratory failure. Crit

Care Med. 2014;42(2):313–21.

93. Ohlow MJ, Moosmann B. Phenothiazine: the seven lives of

pharmacology’s first lead structure. Drug Discov Today.

2011;16(3–4):119–31.

94. Candurra NA, Maskin L, Damonte EB. Inhibition of arenavirus

multiplication in vitro by phenotiazines. Antivir Res.

1996;31(3):149–58.

95. Chu JJ, Ng ML. Infectious entry of West Nile virus occurs

through a clathrin-mediated endocytic pathway. J Virol.

2004;78(19):10543–55.

96. Bhattacharyya S, Warfield KL, Ruthel G, Bavari S, Aman MJ,

Hope TJ. Ebola virus uses clathrin-mediated endocytosis as an

entry pathway. Virology. 2010;401(1):18–28.

97. Chamoun-Emanuelli AM, Pecheur EI, Simeon RL, Huang D,

Cremer PS, Chen Z. Phenothiazines inhibit hepatitis C virus entry,

likely by increasing the fluidity of cholesterol-rich membranes.

Antimicrob Agents Chemother. 2013;57(6):2571–81.

98. Nawa M, Takasaki T, Yamada K, Kurane I, Akatsuka T.

Interference in Japanese encephalitis virus infection of Vero

cells by a cationic amphiphilic drug, chlorpromazine. J Gen

Virol. 2003;84(Pt 7):1737–41.

99. Kaur P, Chu JJ. Chikungunya virus: an update on antiviral

development and challenges. Drug Discov Today.

2013;18(19–20):969–83.

1960 J. Dyall et al.

Page 27: Middle East Respiratory Syndrome and Severe Acute ... · The outbreaks of Middle East respiratory syndrome (MERS) and severe acute respiratory syndrome (SARS) were caused by emerging

100. Johansen LM, DeWald LE, Shoemaker CJ, Hoffstrom BG, Lear-

Rooney CM, Stossel A, et al. A screen of approved drugs and

molecular probes identifies therapeutics with anti-Ebola virus

activity. Sci Transl Med. 2015;7(290):290ra89.

101. Kouznetsova J, Sun W, Martinez-Romero C, Tawa G, Shinn P,

Chen CZ, et al. Identification of 53 compounds that block Ebola

virus-like particle entry via a repurposing screen of approved

drugs. Emerg Microbes Infect. 2014;3(12):e84.

102. Mingorance L, Friesland M, Coto-Llerena M, Perez-del-Pulgar

S, Boix L, Lopez-Oliva JM, et al. Selective inhibition of hep-

atitis C virus infection by hydroxyzine and benztropine.

Antimicrob Agents Chemother. 2014;58(6):3451–60.

103. Carranza M, Snyder MR, Shaw JD, Zesiewicz TA. Parkinson’s

disease. A guide to medical treatment. Turin: SEEd Medical

Publishers; 2013.

104. Cheng H, Lear-Rooney CM, Johansen L, Varhegyi E, Chen ZW,

Olinger GG, et al. Inhibition of Ebola and Marburg virus entry

by G protein-coupled receptor antagonists. J Virol.

2015;89(19):9932–8.

105. Lindquist M, Edwards IR. Risks of non-sedating antihistamines.

Lancet. 1997;349(9061):1322.

106. Chong CR, Chen X, Shi L, Liu JO, Sullivan DJ Jr. A clinical

drug library screen identifies astemizole as an antimalarial

agent. Nat Chem Biol. 2006;2(8):415–6.

107. Garcia-Quiroz J, Camacho J. Astemizole: an old anti-histamine

as a new promising anti-cancer drug. Anticancer Agents Med

Chem. 2011;11(3):307–14.

108. Zhang L, Yu J, Pan H, Hu P, Hao Y, Cai W, et al. Small

molecule regulators of autophagy identified by an image-based

high-throughput screen. Proc Natl Acad Sci USA.

2007;104(48):19023–8.

109. Chiramel AI, Brady NR, Bartenschlager R. Divergent roles of

autophagy in virus infection. Cells. 2013;2(1):83–104.

110. Shoji-Kawata S, Sumpter R, Leveno M, Campbell GR, Zou Z,

Kinch L, et al. Identification of a candidate therapeutic autop-

hagy-inducing peptide. Nature. 2013;494(7436):201–6.

111. Barnard DL, Day CW, Bailey K, Heiner M, Montgomery R,

Lauridsen L, et al. Enhancement of the infectivity of SARS-CoV

in BALB/c mice by IMP dehydrogenase inhibitors, including

ribavirin. Antivir Res. 2006;71(1):53–63.

112. Chan JF, Chan KH, Kao RY, To KK, Zheng BJ, Li CP, et al.

Broad-spectrum antivirals for the emerging Middle East respi-

ratory syndrome coronavirus. J Infect. 2013;67(6):606–16.

113. Cheng KW, Cheng SC, Chen WY, Lin MH, Chuang SJ, Cheng

IH, et al. Thiopurine analogs and mycophenolic acid synergis-

tically inhibit the papain-like protease of Middle East respiratory

syndrome coronavirus. Antivir Res. 2015;115:9–16.

114. Saijo M, Morikawa S, Fukushi S, Mizutani T, Hasegawa H,

Nagata N, et al. Inhibitory effect of mizoribine and ribavirin on

the replication of severe acute respiratory syndrome (SARS)-

associated coronavirus. Antivir Res. 2005;66(2–3):159–63.

115. Cinatl J Jr, Michaelis M, Hoever G, Preiser W, Doerr HW.

Development of antiviral therapy for severe acute respiratory

syndrome. Antivir Res. 2005;66(2–3):81–97.

116. Ibarra KD, Pfeiffer JK. Reduced ribavirin antiviral efficacy via

nucleoside transporter-mediated drug resistance. J Virol.

2009;83(9):4538–47.

117. Smith EC, Denison MR. Coronaviruses as DNA wannabes: a

new model for the regulation of RNA virus replication fidelity.

PLoS Pathog. 2013;9(12):e1003760.

118. Diamond MS, Zachariah M, Harris E. Mycophenolic acid

inhibits dengue virus infection by preventing replication of viral

RNA. Virology. 2002;304(2):211–21.

119. Kitchin JE, Pomeranz MK, Pak G, Washenik K, Shupack JL.

Rediscovering mycophenolic acid: a review of its mechanism,

side effects, and potential uses. J Am Acad Dermatol. 1997;37(3

Pt 1):445–9.

120. Armstrong VW, Tenderich G, Shipkova M, Parsa A, Koerfer R,

Schroder H, et al. Pharmacokinetics and bioavailability of

mycophenolic acid after intravenous administration and oral

administration of mycophenolate mofetil to heart transplant

recipients. Ther Drug Monit. 2005;27(3):315–21.

121. Chan JF, Yao Y, Yeung ML, Deng W, Bao L, Jia L, et al.

Treatment with lopinavir/ritonavir or interferon-beta1b

improves outcome of MERS-CoV Infection in a nonhuman

primate model of common marmoset. J Infect Dis.

2015;212(12):1904–13.

122. Johnson RF, Via LE, Kumar MR, Cornish JP, Yellayi S, Huzella

L, et al. Intratracheal exposure of common marmosets to MERS-

CoV Jordan-n3/2012 or MERS-CoV EMC/2012 isolates does

not result in lethal disease. Virology. 2015;485:422–30.

123. Naka K, Ikeda M, Abe K, Dansako H, Kato N. Mizoribine

inhibits hepatitis C virus RNA replication: effect of combination

with interferon-alpha. Biochem Biophys Res Commun.

2005;330(3):871–9.

124. Clouser CL, Holtz CM, Mullett M, Crankshaw DL, Briggs JE,

Chauhan J, et al. Analysis of the ex vivo and in vivo

antiretroviral activity of gemcitabine. PLoS One.

2011;6(1):e15840.

125. Denisova OV, Kakkola L, Feng L, Stenman J, Nagaraj A,

Lampe J, et al. Obatoclax, saliphenylhalamide, and gemcitabine

inhibit influenza a virus infection. J Biol Chem.

2012;287(42):35324–32.

126. Simmons G, Zmora P, Gierer S, Heurich A, Pohlmann S. Pro-

teolytic activation of the SARS-coronavirus spike protein: cut-

ting enzymes at the cutting edge of antiviral research. Antivir

Res. 2013;100(3):605–14.

127. Glowacka I, Bertram S, Muller MA, Allen P, Soilleux E, Pfef-

ferle S, et al. Evidence that TMPRSS2 activates the severe acute

respiratory syndrome coronavirus spike protein for membrane

fusion and reduces viral control by the humoral immune

response. J Virol. 2011;85(9):4122–34.

128. Matsuyama S, Nagata N, Shirato K, Kawase M, Takeda M,

Taguchi F. Efficient activation of the severe acute respiratory

syndrome coronavirus spike protein by the transmembrane

protease TMPRSS2. J Virol. 2010;84(24):12658–64.

129. Shulla A, Heald-Sargent T, Subramanya G, Zhao J, Perlman S,

Gallagher T. A transmembrane serine protease is linked to the

severe acute respiratory syndrome coronavirus receptor and

activates virus entry. J Virol. 2011;85(2):873–82.

130. Hosoya M, Matsuyama S, Baba M, Suzuki H, Shigeta S. Effects

of protease inhibitors on replication of various myxoviruses.

Antimicrob Agents Chemother. 1992;36(7):1432–6.

131. Kawase M, Shirato K, van der Hoek L, Taguchi F, Matsuyama

S. Simultaneous treatment of human bronchial epithelial cells

with serine and cysteine protease inhibitors prevents severe

acute respiratory syndrome coronavirus entry. J Virol.

2012;86(12):6537–45.

132. Shirato K, Kawase M, Matsuyama S. Middle East respiratory

syndrome coronavirus infection mediated by the transmembrane

serine protease TMPRSS2. J Virol. 2013;87(23):12552–61.

133. Zhou Y, Vedantham P, Lu K, Agudelo J, Carrion R Jr, Nunneley

JW, et al. Protease inhibitors targeting coronavirus and filovirus

entry. Antivir Res. 2015;116:76–84.

134. Chandran K, Sullivan NJ, Felbor U, Whelan SP, Cunningham

JM. Endosomal proteolysis of the Ebola virus glycoprotein is

necessary for infection. Science. 2005;308(5728):1643–5.

135. Schornberg K, Matsuyama S, Kabsch K, Delos S, Bouton A,

White J. Role of endosomal cathepsins in entry mediated by the

Ebola virus glycoprotein. J Virol. 2006;80(8):4174–8.

Therapeutics for MERS-CoV and SARS-CoV 1961

Page 28: Middle East Respiratory Syndrome and Severe Acute ... · The outbreaks of Middle East respiratory syndrome (MERS) and severe acute respiratory syndrome (SARS) were caused by emerging

136. Sham HL, Kempf DJ, Molla A, Marsh KC, Kumar GN, Chen

CM, et al. ABT-378, a highly potent inhibitor of the human

immunodeficiency virus protease. Antimicrob Agents Che-

mother. 1998;42(12):3218–24.

137. Hampson L, Maranga IO, Masinde MS, Oliver AW, Batman G,

He X, et al. A single-arm, proof-of-concept trial of lopimune

(lopinavir/ritonavir) as a treatment for HPV-related pre-invasive

cervical disease. PLoS One. 2016;11(1):e0147917.

138. Wu CY, Jan JT, Ma SH, Kuo CJ, Juan HF, Cheng YS, et al.

Small molecules targeting severe acute respiratory syndrome

human coronavirus. Proc Natl Acad Sci USA.

2004;101(27):10012–7.

139. Cao J, Forrest JC, Zhang X. A screen of the NIH Clinical

Collection small molecule library identifies potential anti-coro-

navirus drugs. Antivir Res. 2015;114:1–10.

140. Sobin BA, Tanner Jr. FW. Anisomycin, a new anti-protozoan

antibiotic. J Am Chem Soc. 1954;76:4053.

141. Akinboye ES, Brennen WN, Rosen DM, Bakare O, Denmeade

SR. Iterative design of emetine-based prodrug targeting fibrob-

last activation protein (FAP) and dipeptidyl peptidase IV DPPIV

using a tandem enzymatic activation strategy. Prostate.

2016;76(8):703–14.

142. Gonzalez Constandse R. Anisomycin in intestinal amebiasis;

study of 30 clinical cases. Prensa Med Mex.

1956;21(7–10):114–5.

143. Conte JE. Manual of antibiotics and infectious diseases: Treat-

ment and prevention. 9th ed. Philadelphia: Lippincott Williams

& Wilkins; 2001.

144. Gupta RS, Krepinsky JJ, Siminovitch L. Structural determinants

responsible for the biological activity of (-)-emetine, (-)-

cryptopleurine, and (-)-tylocrebrine: structure-activity rela-

tionship among related compounds. Mol Pharmacol.

1980;18(1):136–43.

145. Zinck R, Cahill MA, Kracht M, Sachsenmaier C, Hipskind RA,

Nordheim A. Protein synthesis inhibitors reveal differential

regulation of mitogen-activated protein kinase and stress-acti-

vated protein kinase pathways that converge on Elk-1. Mol Cell

Biol. 1995;15(9):4930–8.

146. Ramabhadran TV, Thach RE. Specificity of protein synthesis

inhibitors in the inhibition of encephalomyocarditis virus repli-

cation. J Virol. 1980;34(1):293–6.

147. Hwang YC, Chu JJ, Yang PL, Chen W, Yates MV. Rapid

identification of inhibitors that interfere with poliovirus repli-

cation using a cell-based assay. Antivir Res. 2008;77(3):232–6.

148. Romero MR, Serrano MA, Efferth T, Alvarez M, Marin JJ.

Effect of cantharidin, cephalotaxine and homoharringtonine on

‘‘in vitro’’ models of hepatitis B virus (HBV) and bovine viral

diarrhoea virus (BVDV) replication. Planta Med.

2007;73(6):552–8.

149. Gastaminza P, Whitten-Bauer C, Chisari FV. Unbiased probing

of the entire hepatitis C virus life cycle identifies clinical com-

pounds that target multiple aspects of the infection. Proc Natl

Acad Sci USA. 2010;107(1):291–6.

150. Watashi K, Inoue D, Hijikata M, Goto K, Aly HH, Shimotohno

K. Anti-hepatitis C virus activity of tamoxifen reveals the

functional association of estrogen receptor with viral RNA

polymerase NS5B. J Biol Chem. 2007;282(45):32765–72.

151. Johansen LM, Brannan JM, Delos SE, Shoemaker CJ, Stossel A,

Lear C, et al. FDA-approved selective estrogen receptor mod-

ulators inhibit Ebola virus infection. Sci Transl Med.

2013;5(190):190ra79.

152. Shoemaker CJ, Schornberg KL, Delos SE, Scully C, Pajouhesh

H, Olinger GG, et al. Multiple cationic amphiphiles induce a

Niemann-Pick C phenotype and inhibit Ebola virus entry and

infection. PLoS One. 2013;8(2):e56265.

153. Zhao Y, Ren J, Harlos K, Jones DM, Zeltina A, Bowden TA,

et al. Toremifene interacts with and destabilizes the Ebola virus

glycoprotein. Nature. 2016;535(7610):169–72.

154. Kaufmann AM, Krise JP. Lysosomal sequestration of amine-

containing drugs: analysis and therapeutic implications. J Pharm

Sci. 2007;96(4):729–46.

155. Pharma Orion. FARESTON� (toremifene citrate) tablets pre-

scribing information. Memphis: GTx, Distributor; 2011.

156. AstraZeneca. NOLVADEX� tamoxifen citrate tablets pre-

scribing information. Wilmington: AstraZeneca; 2006.

157. Blanc M, Hsieh WY, Robertson KA, Watterson S, Shui G,

Lacaze P, et al. Host defense against viral infection involves

interferon mediated down-regulation of sterol biosynthesis.

PLoS Biol. 2011;9(3):e1000598.

158. Owens CM, Mawhinney C, Grenier JM, Altmeyer R, Lee MS,

Borisy AA, et al. Chemical combinations elucidate pathway

interactions and regulation relevant to Hepatitis C replication.

Mol Syst Biol. 2010;8(6):375.

159. Warren TK, Jordan R, Lo MK, Ray AS, Mackman RL, Soloveva

V, et al. Therapeutic efficacy of the small molecule GS-5734

against Ebola virus in rhesus monkeys. Nature.

2016;531(7594):381–5.

160. Warren TK, Wells J, Panchal RG, Stuthman KS, Garza NL, Van

Tongeren SA, et al. Protection against filovirus diseases by a

novel broad-spectrum nucleoside analogue BCX4430. Nature.

2014;508(7496):402–5.

161. Peters HL, Jochmans D, de Wilde AH, Posthuma CC, Snijder

EJ, Neyts J, et al. Design, synthesis and evaluation of a series of

acyclic fleximer nucleoside analogues with anti-coronavirus

activity. Bioorg Med Chem Lett. 2015;25(15):2923–6.

162. Adedeji AO, Sarafianos SG. Antiviral drugs specific for coron-

aviruses in preclinical development. Curr Opin Virol.

2014;8:45–53.

163. Tanner JA, Zheng BJ, Zhou J, Watt RM, Jiang JQ, Wong KL,

et al. The adamantane-derived bananins are potent inhibitors of

the helicase activities and replication of SARS coronavirus.

Chem Biol. 2005;12(3):303–11.

164. Leonard JN, Schaffer DV. Antiviral RNAi therapy: emerging

approaches for hitting a moving target. Gene Ther.

2006;13(6):532–40.

165. He ML, Zheng B, Peng Y, Peiris JS, Poon LL, Yuen KY, et al.

Inhibition of SARS-associated coronavirus infection and repli-

cation by RNA interference. JAMA. 2003;290(20):2665–6.

166. He ML, Zheng BJ, Chen Y, Wong KL, Huang JD, Lin MC, et al.

Development of interfering RNA agents to inhibit SARS-asso-

ciated coronavirus infection and replication. Hong Kong Med J.

2009;15(3 Suppl 4):28–31.

167. Lu A, Zhang H, Zhang X, Wang H, Hu Q, Shen L, et al.

Attenuation of SARS coronavirus by a short hairpin RNAexpression plasmid targeting RNA-dependent RNA polymerase.

Virology. 2004;324(1):84–9.

168. Wang Z, Ren L, Zhao X, Hung T, Meng A, Wang J, et al.

Inhibition of severe acute respiratory syndrome virus replication

by small interfering RNAs in mammalian cells. J Virol.

2004;78(14):7523–7.

169. Zheng BJ, Guan Y, Tang Q, Du C, Xie FY, He ML, et al.

Prophylactic and therapeutic effects of small interfering RNA

targeting SARS-coronavirus. Antivir Ther. 2004;9(3):365–74.

170. Zhang Y, Li T, Fu L, Yu C, Li Y, Xu X, et al. Silencing SARS-

CoV spike protein expression in cultured cells by RNA inter-

ference. FEBS Lett. 2004;560(1–3):141–6.

171. Wu CJ, Huang HW, Liu CY, Hong CF, Chan YL. Inhibition of

SARS-CoV replication by siRNA. Antivir Res.

2005;65(1):45–8.

172. Akerstrom S, Mirazimi A, Tan YJ. Inhibition of SARS-CoV

replication cycle by small interference RNAs silencing specific

1962 J. Dyall et al.

Page 29: Middle East Respiratory Syndrome and Severe Acute ... · The outbreaks of Middle East respiratory syndrome (MERS) and severe acute respiratory syndrome (SARS) were caused by emerging

SARS proteins, 7a/7b, 3a/3b and S. Antivir Res.

2007;73(3):219–27.

173. Li BJ, Tang Q, Cheng D, Qin C, Xie FY, Wei Q, et al. Using

siRNA in prophylactic and therapeutic regimens against SARS

coronavirus in rhesus macaque. Nat Med. 2005;11(9):944–51.

174. Hasan MM, Akter R, Ullah MS, Abedin MJ, Ullah GM, Hossain

MZ. A computational approach for predicting role of human

microRNAs in MERS-CoV genome. Adv Bioinform.

2014;2014:967946.

175. Thi EP, Mire CE, Lee AC, Geisbert JB, Zhou JZ, Agans KN,

et al. Lipid nanoparticle siRNA treatment of Ebola-virus-

Makona-infected nonhuman primates. Nature.

2015;521(7552):362–5.

176. Dunning J, Sahr F, Rojek A, Gannon F, Carson G, Idriss B, et al.

Experimental treatment of Ebola virus disease with TKM-

130803: a single-arm phase 2 clinical trial. PLoS Med.

2016;13(4):e1001997.

177. Badani H, Garry RF, Wimley WC. Peptide entry inhibitors of

enveloped viruses: the importance of interfacial hydrophobicity.

Biochim Biophys Acta. 2014;1838(9):2180–97.

178. Lu L, Liu Q, Zhu Y, Chan KH, Qin L, Li Y, et al. Structure-

based discovery of Middle East respiratory syndrome coron-

avirus fusion inhibitor. Nat Commun. 2014;5:3067.

179. Sainz B Jr, Mossel EC, Gallaher WR, Wimley WC, Peters CJ,

Wilson RB, et al. Inhibition of severe acute respiratory syn-

drome-associated coronavirus (SARS-CoV) infectivity by pep-

tides analogous to the viral spike protein. Virus Res.

2006;120(1–2):146–55.

180. Sainz B Jr, Rausch JM, Gallaher WR, Garry RF, Wimley WC.

Identification and characterization of the putative fusion peptide

of the severe acute respiratory syndrome-associated coronavirus

spike protein. J Virol. 2005;79(11):7195–206.

181. Bird GH, Madani N, Perry AF, Princiotto AM, Supko JG, He X,

et al. Hydrocarbon double-stapling remedies the proteolytic

instability of a lengthy peptide therapeutic. Proc Natl Acad Sci

USA. 2010;107(32):14093–8.

182. Kindrachuk J, Scruten E, Attah-Poku S, Bell K, Potter A,

Babiuk LA, et al. Stability, toxicity, and biological activity of

host defense peptide BMAP28 and its inversed and retro-in-

versed isomers. Biopolymers. 2011;96(1):14–24.

183. Walensky LD, Bird GH. Hydrocarbon-stapled peptides: princi-

ples, practice, and progress. J Med Chem. 2014;57(15):6275–88.

184. Shadman KA, Wald ER. A review of palivizumab and emerging

therapies for respiratory syncytial virus. Expert Opin Biol Ther.

2011;11(11):1455–67.

185. Qiu X, Wong G, Audet J, Bello A, Fernando L, Alimonti JB,

et al. Reversion of advanced Ebola virus disease in nonhuman

primates with ZMapp. Nature. 2014;514(7520):47–53.

186. PREVAIL II Writing Group for the Multinational PREVAIL

Study Team. A randomized, controlled trial of ZMapp for Ebola

virus fnfection. N Engl J Med. 2016;375(15):1448–56.

187. Bossart KN, Zhu Z, Middleton D, Klippel J, Crameri G, Bing-

ham J, et al. A neutralizing human monoclonal antibody protects

against lethal disease in a new ferret model of acute nipah virus

infection. PLoS Pathog. 2009;5(10):e1000642.

188. Zhu Z, Dimitrov AS, Bossart KN, Crameri G, Bishop KA,

Choudhry V, et al. Potent neutralization of Hendra and Nipah

viruses by human monoclonal antibodies. J Virol.

2006;80(2):891–9.

189. Bossart KN, Geisbert TW, Feldmann H, Zhu Z, Feldmann F,

Geisbert JB, et al. A neutralizing human monoclonal antibody

protects African green monkeys from hendra virus challenge.

Sci Transl Med. 2011;3(105):105ra3.

190. Corti D, Zhao J, Pedotti M, Simonelli L, Agnihothram S, Fett C,

et al. Prophylactic and postexposure efficacy of a potent human

monoclonal antibody against MERS coronavirus. Proc Natl

Acad Sci USA. 2015;112(33):10473–8.

191. Houser KV, Gretebeck L, Ying T, Wang Y, Vogel L, Lamirande

EW, et al. Prophylaxis with a Middle East respiratory syndrome

coronavirus (MERS-CoV)-specific human monoclonal antibody

protects rabbits from MERS-CoV infection. J Infect Dis.

2016;213(10):1557–61.

192. Jiang L, Wang N, Zuo T, Shi X, Poon KM, Wu Y, et al. Potent

neutralization of MERS-CoV by human neutralizing mono-

clonal antibodies to the viral spike glycoprotein. Sci Transl Med.

2014;6(234):234ra59.

193. Johnson RF, Bagci U, Keith L, Tang X, Mollura DJ, Zeitlin L,

et al. 3B11-N, a monoclonal antibody against MERS-CoV,

reduces lung pathology in rhesus monkeys following intratra-

cheal inoculation of MERS-CoV Jordan-n3/2012. Virology.

2016;490:49–58.

194. Pascal KE, Coleman CM, Mujica AO, Kamat V, Badithe A,

Fairhurst J, et al. Pre- and postexposure efficacy of fully human

antibodies against Spike protein in a novel humanized mouse

model of MERS-CoV infection. Proc Natl Acad Sci USA.

2015;112(28):8738–43.

195. Qiu H, Sun S, Xiao H, Feng J, Guo Y, Tai W, et al. Single-dose

treatment with a humanized neutralizing antibody affords full

protection of a human transgenic mouse model from lethal

Middle East respiratory syndrome (MERS)-coronavirus infec-

tion. Antivir Res. 2016;14(132):141–8.

196. Tang XC, Agnihothram SS, Jiao Y, Stanhope J, Graham RL,

Peterson EC, et al. Identification of human neutralizing anti-

bodies against MERS-CoV and their role in virus adaptive

evolution. Proc Natl Acad Sci USA. 2014;111(19):E2018–26.

197. Ying T, Du L, Ju TW, Prabakaran P, Lau CC, Lu L, et al.

Exceptionally potent neutralization of Middle East respiratory

syndrome coronavirus by human monoclonal antibodies. J Virol.

2014;88(14):7796–805.

198. Mair-Jenkins J, Saavedra-Campos M, Baillie JK, Cleary P,

Khaw FM, Lim WS, et al. The effectiveness of convalescent

plasma and hyperimmune immunoglobulin for the treatment of

severe acute respiratory infections of viral etiology: a systematic

review and exploratory meta-analysis. J Infect Dis.

2015;211(1):80–90.

199. Ying T, Li H, Lu L, Dimitrov DS, Jiang S. Development of

human neutralizing monoclonal antibodies for prevention and

therapy of MERS-CoV infections. Microbes Infect.

2015;17(2):142–8.

200. Berry JD, Gaudet RG. Antibodies in infectious diseases: poly-

clonals, monoclonals and niche biotechnology. New Biotechnol.

2011;28(5):489–501.

201. Luke T, Wu H, Zhao J, Channappanavar R, Coleman CM, Jiao

JA, et al. Human polyclonal immunoglobulin G from tran-

schromosomic bovines inhibits MERS-CoV in vivo. Sci Transl

Med. 2016;8(326):326ra21.

202. De Clercq E. Strategies in the design of antiviral drugs. Nat Rev

Drug Discov. 2002;1(1):13–25.

203. AIDSinfo. HIV treatment. FDA-approved HIV medicines. 2016.

https://aidsinfo.nih.gov/education-materials/fact-sheets/21/58/

fda-approved-hiv-medicines. Accessed 13 Oct 2016.

204. World Health Organization. Consolidated guidelines for the use

of antiretroviral drugs for treating and preventing HIV infection.

Recommendations for a public health approach, 2nd edn. 2016.

http://www.who.int/hiv/pub/arv/arv-2016/en/. Accessed 13 Oct

2016.

205. American Association for the Study of Liver Diseases, Infec-

tious Diseases Society of America. HCV guidance: recommen-

dations for testing, managing, and treating hepatitis C. 2017.

http://www.hcvguidelines.org./sites/default/files/full-guidance-

Therapeutics for MERS-CoV and SARS-CoV 1963

Page 30: Middle East Respiratory Syndrome and Severe Acute ... · The outbreaks of Middle East respiratory syndrome (MERS) and severe acute respiratory syndrome (SARS) were caused by emerging

pdf/HCVGuidance_April_12_2017_b.pdf. Accessed 28 July

2017.

206. Kohli A, Shaffer A, Sherman A, Kottilil S. Treatment of hep-

atitis C: a systematic review. JAMA. 2014;312(6):631–40.

207. Ogawa E, Furusyo N, Kajiwara E, Takahashi K, Nomura H,

Tanabe Y, et al. Evaluation of the adverse effect of premature

discontinuation of pegylated interferon alpha-2b and ribavirin

treatment for chronic hepatitis C virus infection: results from

Kyushu University liver disease study. J Gastroenterol Hepatol.

2012;27(7):1233–40.

208. Govorkova EA, McCullers JA. Therapeutics against influenza.

Curr Top Microbiol Immunol. 2013;370:273–300.

209. Dunning J, Baillie JK, Cao B, Hayden FG. International Severe

Acute Respiratory Emerging Infection Consortium. Antiviral

combinations for severe influenza. Lancet Infect Dis.

2014;14(12):1259–70.

210. Hayden FG. Advances in antivirals for non-influenza respiratory

virus infections. Influenza Other Respir Viruses. 2013;7(Suppl

3):36–43.

211. Seo S, Englund JA, Nguyen JT, Pukrittayakamee S, Lindegardh

N, Tarning J, et al. Combination therapy with amantadine,

oseltamivir and ribavirin for influenza A infection: safety and

pharmacokinetics. Antivir Ther. 2013;18(3):377–86.

212. Agnandji ST, Huttner A, Zinser ME, Njuguna P, Dahlke C,

Fernandes JF, et al. Phase 1 Trials of rVSV Ebola Vaccine in

Africa and Europe. N Engl J Med. 2016;374(17):1647–60.

213. Ewer K, Rampling T, Venkatraman N, Bowyer G, Wright D,

Lambe T, et al. A monovalent chimpanzee adenovirus Ebola

vaccine boosted with MVA. N Engl J Med.

2016;374(17):1635–46.

214. Ledgerwood JE, DeZure AD, Stanley DA, Coates EE, Novik L,

Enama ME, et al. Chimpanzee adenovirus vector Ebola vaccine.

N Engl J Med. 2017; 376(10):928–38.

215. Regules JA, Beigel JH, Paolino KM, Voell J, Castellano AR, Hu

Z, et al. A recombinant vesicular stomatitis virus Ebola vaccine.

N Engl J Med. 2017;376(4):330–41.

216. Henao-Restrepo AM, Longini IM, Egger M, Dean NE, Edmunds

WJ, Camacho A, et al. Efficacy and effectiveness of an rVSV-

vectored vaccine expressing Ebola surface glycoprotein: interim

results from the Guinea ring vaccination cluster-randomised

trial. Lancet. 2015;386(9996):857–66.

217. Baseler L, de Wit E, Feldmann H. A comparative review of

animal models of Middle East respiratory syndrome coronavirus

infection. Vet Pathol. 2016;53(3):521–31.

218. Sutton TC, Subbarao K. Development of animal models against

emerging coronaviruses: from SARS to MERS coronavirus.

Virology. 2015;479–480:247–58.

219. van Doremalen N, Munster VJ. Animal models of Middle East

respiratory syndrome coronavirus infection. Antivir Res.

2015;122:28–38.

220. Zhao J, Li K, Wohlford-Lenane C, Agnihothram SS, Fett C,

Zhao J, et al. Rapid generation of a mouse model for Middle

East respiratory syndrome. Proc Natl Acad Sci USA.

2014;111(13):4970–5.

221. Channappanavar R, Fett C, Zhao J, Meyerholz DK, Perlman S.

Virus-specific memory CD8 T cells provide substantial protec-

tion from lethal severe acute respiratory syndrome coronavirus

infection. J Virol. 2014;88(19):11034–44.

222. Channappanavar R, Zhao J, Perlman S. T cell-mediated immune

response to respiratory coronaviruses. Immunol Res.

2014;59(1–3):118–28.

223. Agrawal AS, Garron T, Tao X, Peng BH, Wakamiya M, Chan

TS, et al. Generation of a transgenic mouse model of Middle

East respiratory syndrome coronavirus infection and disease.

J Virol. 2015;89(7):3659–70.

224. Cockrell AS, Yount BL, Scobey T, Jensen K, Douglas M, Beall

A, et al. A mouse model for MERS coronavirus-induced acute

respiratory distress syndrome. Nat Microbiol. 2016;28(2):16226.

225. Haagmans BL, van den Brand JM, Provacia LB, Raj VS, Stit-

telaar KJ, Getu S, et al. Asymptomatic Middle East respiratory

syndrome coronavirus infection in rabbits. J Virol.

2015;89(11):6131–5.

226. de Wit E, Rasmussen AL, Falzarano D, Bushmaker T, Feldmann

F, Brining DL, et al. Middle East respiratory syndrome coron-

avirus (MERS-CoV) causes transient lower respiratory tract

infection in rhesus macaques. Proc Natl Acad Sci USA.

2013;110(41):16598–603.

227. Munster VJ, de Wit E, Feldmann H. Pneumonia from human

coronavirus in a macaque model. N Engl J Med.

2013;368(16):1560–2.

228. Yao Y, Bao L, Deng W, Xu L, Li F, Lv Q, et al. An animal

model of MERS produced by infection of rhesus macaques with

MERS coronavirus. J Infect Dis. 2014;209(2):236–42.

229. Clay CC, Donart N, Fomukong N, Knight JB, Overheim K,

Tipper J, et al. Severe acute respiratory syndrome-coronavirus

infection in aged nonhuman primates is associated with modu-

lated pulmonary and systemic immune responses. Immun Age-

ing. 2014;11(1):4.

230. Smits SL, van den Brand JM, de Lang A, Leijten LM, van Ijcken

WF, van Amerongen G, et al. Distinct severe acute respiratory

syndrome coronavirus-induced acute lung injury pathways in

two different nonhuman primate species. J Virol.

2011;85(9):4234–45.

231. McAuliffe J, Vogel L, Roberts A, Fahle G, Fischer S, Shieh WJ,

et al. Replication of SARS coronavirus administered into the

respiratory tract of African green, rhesus and cynomolgus

monkeys. Virology. 2004;330(1):8–15.

232. Hartman AL, Powell DS, Bethel LM, Caroline AL, Schmid RJ,

Oury T, et al. Aerosolized Rift Valley fever virus causes fatal

encephalitis in African green monkeys and common marmosets.

J Virol. 2014;88(4):2235–45.

233. Kramski M, Matz-Rensing K, Stahl-Hennig C, Kaup FJ, Nitsche

A, Pauli G, et al. A novel highly reproducible and lethal non-

human primate model for orthopox virus infection. PLoS One.

2010;5(4):e10412.

234. Mucker EM, Chapman J, Huzella LM, Huggins JW, Shamblin J,

Robinson CG, et al. Susceptibility of marmosets (Callithrix

jacchus) to monkeypox virus: a low dose prospective model for

monkeypox and smallpox disease. PLoS One.

2015;10(7):e0131742.

235. Falzarano D, de Wit E, Feldmann F, Rasmussen AL, Okumura

A, Peng X, et al. Infection with MERS-CoV causes lethal

pneumonia in the common marmoset. PLoS Pathog.

2014;10(8):e1004250.

236. Greenough TC, Carville A, Coderre J, Somasundaran M, Sul-

livan JL, Luzuriaga K, et al. Pneumonitis and multi-organ sys-

tem disease in common marmosets (Callithrix jacchus) infected

with the severe acute respiratory syndrome-associated coron-

avirus. Am J Pathol. 2005;167(2):455–63.

237. Bai JP. Pharmacodynamics and systems pharmacology approa-

ches to repurposing drugs in the wake of global health burden.

J Pharm Sci. 2016;105(10):3007–12.

238. Brik A, Wong CH. HIV-1 protease: mechanism and drug dis-

covery. Org Biomol Chem. 2003;1(1):5–14.

239. Navia MA, Fitzgerald PM, McKeever BM, Leu CT, Heimbach

JC, Herber WK, et al. Three-dimensional structure of aspartyl

protease from human immunodeficiency virus HIV-1. Nature.

1989;337(6208):615–20.

240. Wlodawer A, Miller M, Jaskolski M, Sathyanarayana BK,

Baldwin E, Weber IT, et al. Conserved folding in retroviral

1964 J. Dyall et al.

Page 31: Middle East Respiratory Syndrome and Severe Acute ... · The outbreaks of Middle East respiratory syndrome (MERS) and severe acute respiratory syndrome (SARS) were caused by emerging

proteases: crystal structure of a synthetic HIV-1 protease. Sci-

ence. 1989;245(4918):616–21.

241. Gotte M, Feld JJ. Direct-acting antiviral agents for hepatitis C:

structural and mechanistic insights. Nat Rev Gastroenterol

Hepatol. 2016;13(6):338–51.

242. Hazuda DJ, Burroughs M, Howe AY, Wahl J, Venkatraman S.

Development of boceprevir: a first-in-class direct antiviral

treatment for chronic hepatitis C infection. Ann N Y Acad Sci.

2013;1291:69–76.

243. Kwong AD, Kauffman RS, Hurter P, Mueller P. Discovery and

development of telaprevir: an NS3-4A protease inhibitor for

treating genotype 1 chronic hepatitis C virus. Nat Biotechnol.

2011;29(11):993–1003.

244. Hilgenfeld R. From SARS to MERS: crystallographic studies on

coronaviral proteases enable antiviral drug design. FEBS J.

2014;281(18):4085–96.

245. Miller ME, Adhikary S, Kolokoltsov AA, Davey RA. Ebola-

virus requires acid sphingomyelinase activity and plasma

membrane sphingomyelin for infection. J Virol.

2012;86(14):7473–83.

246. Muller MP, Dresser L, Raboud J, McGeer A, Rea E, Richardson

SE, et al. Adverse events associated with high-dose ribavirin:

evidence from the Toronto outbreak of severe acute respiratory

syndrome. Pharmacotherapy. 2007;27(4):494–503.

247. Ward SE, Loutfy MR, Blatt LM, Siminovitch KA, Chen J,

Hinek A, et al. Dynamic changes in clinical features and cyto-

kine/chemokine responses in SARS patients treated with inter-

feron alfacon-1 plus corticosteroids. Antivir Ther.

2005;10(2):263–75.

248. Zhao Z, Zhang F, Xu M, Huang K, Zhong W, Cai W, et al.

Description and clinical treatment of an early outbreak of severe

acute respiratory syndrome (SARS) in Guangzhou, PR China.

J Med Microbiol. 2003;52(Pt 8):715–20.

249. Al-Tawfiq JA, Momattin H, Dib J, Memish ZA. Ribavirin and

interferon therapy in patients infected with the Middle East

respiratory syndrome coronavirus: an observational study. Int J

Infect Dis. 2014;20:42–6.

250. Boonyasuppayakorn S, Reichert ED, Manzano M, Nagarajan K,

Padmanabhan R. Amodiaquine, an antimalarial drug, inhibits

dengue virus type 2 replication and infectivity. Antivir Res.

2014;106:125–34.

251. Bassetto M, De Burghgraeve T, Delang L, Massarotti A,

Coluccia A, Zonta N, et al. Computer-aided identification,

design and synthesis of a novel series of compounds with

selective antiviral activity against chikungunya virus. Antivir

Res. 2013;98(1):12–8.

252. Cruz DJ, Bonotto RM, Gomes RG, da Silva CT, Taniguchi JB,

No JH, et al. Identification of novel compounds inhibiting

chikungunya virus-induced cell death by high throughput

screening of a kinase inhibitor library. PLoS Negl Trop Dis.

2013;7(10):e2471.

253. Yan Y, Zou Z, Sun Y, Li X, Xu KF, Wei Y, et al. Anti-malaria

drug chloroquine is highly effective in treating avian influenza A

H5N1 virus infection in an animal model. Cell Res.

2013;23(2):300–2.

254. Paton NI, Lee L, Xu Y, Ooi EE, Cheung YB, Archuleta S, et al.

Chloroquine for influenza prevention: a randomised, double-

blind, placebo controlled trial. Lancet Infect Dis.

2011;11(9):677–83.

255. Romanelli F, Smith KM, Hoven AD. Chloroquine and hydrox-

ychloroquine as inhibitors of human immunodeficiency virus

(HIV-1) activity. Curr Pharm Des. 2004;10(21):2643–8.

256. Nakagawa M, Sakamoto N, Tanabe Y, Koyama T, Itsui Y,

Takeda Y, et al. Suppression of hepatitis C virus replication by

cyclosporin A is mediated by blockade of cyclophilins. Gas-

troenterology. 2005;129(3):1031–41.

257. Qing M, Yang F, Zhang B, Zou G, Robida JM, Yuan Z, et al.

Cyclosporine inhibits flavivirus replication through blocking the

interaction between host cyclophilins and viral NS5 protein.

Antimicrob Agents Chemother. 2009;53(8):3226–35.

258. Kambara H, Tani H, Mori Y, Abe T, Katoh H, Fukuhara T, et al.

Involvement of cyclophilin B in the replication of Japanese

encephalitis virus. Virology. 2011;412(1):211–9.

259. Bose S, Mathur M, Bates P, Joshi N, Banerjee AK. Requirement

for cyclophilin A for the replication of vesicular stomatitis virus

New Jersey serotype. J Gen Virol. 2003;84(Pt 7):1687–99.

260. Briggs CJ, Ott DE, Coren LV, Oroszlan S, Tozser J. Comparison

of the effect of FK506 and cyclosporin A on virus production in

H9 cells chronically and newly infected by HIV-1. Arch Virol.

1999;144(11):2151–60.

261. Randhawa PS, Farasati NA, Huang Y, Mapara MY, Shapiro R.

Viral drug sensitivity testing using quantitative PCR: effect of

tyrosine kinase inhibitors on polyomavirus BK replication. Am J

Clin Pathol. 2010;134(6):916–20.

262. Pogliaghi M, Papagno L, Lambert S, Calin R, Calvez V, Kat-

lama C, et al. The tyrosine kinase inhibitor Dasatinib blocks in-

vitro HIV-1 production by primary CD4? T cells from HIV-1

infected patients. AIDS. 2014;28(2):278–81.

263. de Wispelaere M, LaCroix AJ, Yang PL. The small molecules

AZD0530 and dasatinib inhibit dengue virus RNA replication

via Fyn kinase. J Virol. 2013;87(13):7367–81.

264. Reeves PM, Smith SK, Olson VA, Thorne SH, Bornmann W,

Damon IK, et al. Variola and monkeypox viruses utilize con-

served mechanisms of virion motility and release that depend on

ABL and SRC family tyrosine kinases. J Virol.

2011;85(1):21–31.

265. Murray JL, McDonald NJ, Sheng J, Shaw MW, Hodge TW,

Rubin DH, et al. Inhibition of influenza A virus replication by

antagonism of a PI3 K-AKT-mTOR pathway member identified

by gene-trap insertional mutagenesis. Antivir Chem Chemother.

2012;22(5):205–15.

266. Brennan DC, Legendre C, Patel D, Mange K, Wiland A,

McCague K, et al. Cytomegalovirus incidence between ever-

olimus versus mycophenolate in de novo renal transplants:

pooled analysis of three clinical trials. Am J Transplant.

2011;11(11):2453–62.

267. Kobashigawa J, Ross H, Bara C, Delgado JF, Dengler T,

Lehmkuhl HB, et al. Everolimus is associated with a reduced

incidence of cytomegalovirus infection following de novo car-

diac transplantation. Transpl Infect Dis. 2013;15(2):150–62.

268. Hutterer C, Wandinger SK, Wagner S, Muller R, Stamminger T,

Zeittrager I, et al. Profiling of the kinome of cytomegalovirus-

infected cells reveals the functional importance of host kinases

Aurora A, ABL and AMPK. Antivir Res. 2013;99(2):139–48.

269. Cheshenko N, Trepanier JB, Stefanidou M, Buckley N, Gon-

zalez P, Jacobs W, et al. HSV activates Akt to trigger calcium

release and promote viral entry: novel candidate target for

treatment and suppression. FASEB J. 2013;27(7):2584–99.

270. Heredia A, Gilliam B, Latinovic O, Le N, Bamba D, Devico A,et al. Rapamycin reduces CCR5 density levels on CD4 T cells,

and this effect results in potentiation of enfuvirtide (T-20)

against R5 strains of human immunodeficiency virus type 1

in vitro. Antimicrob Agents Chemother. 2007;51(7):2489–96.

271. Kudchodkar SB, Yu Y, Maguire TG, Alwine JC. Human cyto-

megalovirus infection alters the substrate specificities and

rapamycin sensitivities of raptor- and rictor-containing com-

plexes. Proc Natl Acad Sci USA. 2006;103(38):14182–7.

272. Di Benedetto F, Di Sandro S, De Ruvo N, Montalti R, Ballarin

R, Guerrini GP, et al. First report on a series of HIV patients

undergoing rapamycin monotherapy after liver transplantation.

Transplantation. 2010;89(6):733–8.

Therapeutics for MERS-CoV and SARS-CoV 1965

Page 32: Middle East Respiratory Syndrome and Severe Acute ... · The outbreaks of Middle East respiratory syndrome (MERS) and severe acute respiratory syndrome (SARS) were caused by emerging

273. Ghassemieh B, Ahya VN, Baz MA, Valentine VG, Arcasoy SM,

Love RB, et al. Decreased incidence of cytomegalovirus infec-

tion with sirolimus in a post hoc randomized, multicenter study

in lung transplantation. J Heart Lung Transplant.

2013;32(7):701–6.

274. Soliman A, Fathy A, Khashab S, Shaheen N, Soliman M. Sir-

olimus conversion may suppress viral replication in hepatitis C

virus-positive renal transplant candidates. Exp Clin Transplant.

2013;11(5):408–11.

275. Michaelis M, Paulus C, Loschmann N, Dauth S, Stange E, Doerr

HW, et al. The multi-targeted kinase inhibitor sorafenib inhibits

human cytomegalovirus replication. Cell Mol Life Sci.

2011;68(6):1079–90.

276. Gao M, Duan H, Liu J, Zhang H, Wang X, Zhu M, et al. The

multi-targeted kinase inhibitor sorafenib inhibits enterovirus 71

replication by regulating IRES-dependent translation of viral

proteins. Antivir Res. 2014;106:80–5.

277. Benedict A, Bansal N, Senina S, Hooper I, Lundberg L, de la

Fuente C, et al. Repurposing FDA-approved drugs as thera-

peutics to treat Rift Valley fever virus infection. Front Micro-

biol. 2015;6:676.

278. Sasaki H, Nakamura M, Ohno T, Matsuda Y, Yuda Y, Nono-

mura Y. Myosin-actin interaction plays an important role in

human immunodeficiency virus type 1 release from host cells.

Proc Natl Acad Sci USA. 1995;92(6):2026–30.

279. Hirai H, Takeda S, Natori S, Sekimizu K. Inhibition of SV40

DNA replication in vitro by chlorpromazine. Biol Pharm Bull.

1993;16(6):565–7.

280. Yanagida K, Baba C, Baba M. Inhibition of bovine viral diar-

rhea virus (BVDV) by mizoribine: synergistic effect of combi-

nation with interferon-alpha. Antivir Res. 2004;64(3):195–201.

281. Batman G, Oliver AW, Zehbe I, Richard C, Hampson L,

Hampson IN. Lopinavir up-regulates expression of the antiviral

protein ribonuclease L in human papillomavirus-positive cervi-

cal carcinoma cells. Antivir Ther. 2011;16(4):515–25.

282. Murakami Y, Fukasawa M, Kaneko Y, Suzuki T, Wakita T,

Fukazawa H. Selective estrogen receptor modulators inhibit

hepatitis C virus infection at multiple steps of the virus life

cycle. Microbes Infect. 2013;15(1):45–55.

283. Zheng K, Chen M, Xiang Y, Ma K, Jin F, Wang X, et al.

Inhibition of herpes simplex virus type 1 entry by chloride

channel inhibitors tamoxifen and NPPB. Biochem Biophys Res

Commun. 2014;446(4):990–6.

284. Kilianski A, Mielech AM, Deng X, Baker SC. Assessing activity

and inhibition of Middle East respiratory syndrome coronavirus

papain-like and 3C-like proteases using luciferase-based

biosensors. J Virol. 2013;87(21):11955–62.

285. Lundin A, Dijkman R, Bergstrom T, Kann N, Adamiak B,

Hannoun C, et al. Targeting membrane-bound viral RNA syn-

thesis reveals potent inhibition of diverse coronaviruses

including the Middle East respiratory syndrome virus. PLoS

Pathog. 2014;10(5):e1004166.

286. Millet JK, Whittaker GR. Host cell entry of Middle East res-

piratory syndrome coronavirus after two-step, furin-mediated

activation of the spike protein. Proc Natl Acad Sci USA.

2014;111(42):15214–9.

287. Shie JJ, Fang JM, Kuo CJ, Kuo TH, Liang PH, Huang HJ, et al.

Discovery of potent anilide inhibitors against the severe acute

respiratory syndrome 3CL protease. J Med Chem.

2005;48(13):4469–73.

288. Shao YM, Yang WB, Peng HP, Hsu MF, Tsai KC, Kuo TH,

et al. Structure-based design and synthesis of highly potent

SARS-CoV 3CL protease inhibitors. ChemBioChem.

2007;8(14):1654–7.

289. Ramajayam R, Tan KP, Liu HG, Liang PH. Synthesis and

evaluation of pyrazolone compounds as SARS-coronavirus 3C-

like protease inhibitors. Bioorg Med Chem.

2010;18(22):7849–54.

290. Shao YM, Yang WB, Kuo TH, Tsai KC, Lin CH, Yang AS,

et al. Design, synthesis, and evaluation of trifluoromethyl

ketones as inhibitors of SARS-CoV 3CL protease. Bioorg Med

Chem. 2008;16(8):4652–60.

291. Chen L, Gui C, Luo X, Yang Q, Gunther S, Scandella E, et al.

Cinanserin is an inhibitor of the 3C-like proteinase of severe

acute respiratory syndrome coronavirus and strongly reduces

virus replication in vitro. J Virol. 2005;79(11):7095–103.

292. Lee CC, Kuo CJ, Hsu MF, Liang PH, Fang JM, Shie JJ, et al.

Structural basis of mercury- and zinc-conjugated complexes as

SARS-CoV 3C-like protease inhibitors. FEBS Lett.

2007;581(28):5454–8.

293. Lee C, Lee JM, Lee NR, Kim DE, Jeong YJ, Chong Y. Inves-

tigation of the pharmacophore space of severe acute respiratory

syndrome coronavirus (SARS-CoV) NTPase/helicase by dihy-

droxychromone derivatives. Bioorg Med Chem Lett.

2009;19(16):4538–41.

294. Kim MK, Yu MS, Park HR, Kim KB, Lee C, Cho SY, et al. 2,6-

Bis-arylmethyloxy-5-hydroxychromones with antiviral activity

against both hepatitis C virus (HCV) and SARS-associated

coronavirus (SCV). Eur J Med Chem. 2011;46(11):5698–704.

295. Cho A, Saunders OL, Butler T, Zhang L, Xu J, Vela JE, et al.

Synthesis and antiviral activity of a series of 1’-substituted

4-aza-7,9-dideazaadenosine C-nucleosides. Bioorg Med Chem

Lett. 2012;22(8):2705–7.

296. Shah PP, Wang T, Kaletsky RL, Myers MC, Purvis JE, Jing H,

et al. A small-molecule oxocarbazate inhibitor of human

cathepsin L blocks severe acute respiratory syndrome and Ebola

pseudotype virus infection into human embryonic kidney 293T

cells. Mol Pharmacol. 2010;78(2):319–24.

297. Adedeji AO, Severson W, Jonsson C, Singh K, Weiss SR,

Sarafianos SG. Novel inhibitors of severe acute respiratory

syndrome coronavirus entry that act by three distinct mecha-

nisms. J Virol. 2013;87(14):8017–28.

298. Huentelman MJ, Zubcevic J, Hernandez Prada JA, Xiao X,

Dimitrov DS, Raizada MK, et al. Structure-based discovery of a

novel angiotensin-converting enzyme 2 inhibitor. Hypertension.

2004;44(6):903–6.

1966 J. Dyall et al.